Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
FASEB J ; 29(2): 684-95, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25392266

RESUMO

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.


Assuntos
Sistema Nervoso Entérico/fisiologia , Trato Gastrointestinal/inervação , Sistema Imunitário/fisiologia , Lacticaseibacillus rhamnosus/imunologia , Animais , Células da Medula Óssea/citologia , Linfócitos T CD4-Positivos/citologia , Chaperonina 60/metabolismo , Técnicas de Cocultura , Células Dendríticas/citologia , Células Dendríticas/microbiologia , Fatores de Transcrição Forkhead/metabolismo , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Lectinas Tipo C/metabolismo , Linfonodos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neurônios/metabolismo , Peristaltismo , Nódulos Linfáticos Agregados/microbiologia , Fenótipo , Probióticos , Proteômica , Transdução de Sinais
2.
J Physiol ; 593(17): 3943-57, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26084409

RESUMO

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.


Assuntos
Bradicardia/terapia , Jejuno/fisiologia , Limosilactobacillus reuteri , Probióticos , Gastropatias/terapia , Canais de Cátion TRPV/fisiologia , Analgesia , Animais , Bradicardia/etiologia , Bradicardia/fisiopatologia , Capsaicina , Gânglios Espinais/fisiologia , Jejuno/inervação , Masculino , Mesentério/inervação , Mesentério/fisiologia , Camundongos Knockout , Probióticos/farmacologia , Probióticos/uso terapêutico , Ratos Sprague-Dawley , Nervos Espinhais/fisiologia , Gastropatias/complicações , Gastropatias/fisiopatologia , Canais de Cátion TRPV/genética
3.
FASEB J ; 28(7): 3064-74, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24719355

RESUMO

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."


Assuntos
Encéfalo/fisiologia , Jejuno/fisiologia , Neurônios Aferentes/fisiologia , Receptores Nicotínicos/metabolismo , Sinapses/metabolismo , Nervo Vago/fisiologia , Potenciais de Ação/fisiologia , Animais , Encéfalo/metabolismo , Jejuno/inervação , Jejuno/metabolismo , Jejuno/microbiologia , Lacticaseibacillus rhamnosus/metabolismo , Masculino , Camundongos , Nervo Vago/metabolismo
4.
Commun Biol ; 7(1): 80, 2024 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-38200107

RESUMO

Vagus nerve signaling is a key component of the gut-brain axis and regulates diverse physiological processes that decline with age. Gut to brain vagus firing patterns are regulated by myenteric intrinsic primary afferent neuron (IPAN) to vagus neurotransmission. It remains unclear how IPANs or the afferent vagus age functionally. Here we identified a distinct ageing code in gut to brain neurotransmission defined by consistent differences in firing rates, burst durations, interburst and intraburst firing intervals of IPANs and the vagus, when comparing young and aged neurons. The aminosterol squalamine changed aged neurons firing patterns to a young phenotype. In contrast to young neurons, sertraline failed to increase firing rates in the aged vagus whereas squalamine was effective. These results may have implications for improved treatments involving pharmacological and electrical stimulation of the vagus for age-related mood and other disorders. For example, oral squalamine might be substituted for or added to sertraline for the aged.


Assuntos
Células Receptoras Sensoriais , Sertralina , Colestanóis , Nervo Vago
5.
Am J Physiol Gastrointest Liver Physiol ; 304(2): G211-20, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23139216

RESUMO

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.


Assuntos
Sistema Nervoso Entérico/fisiologia , Jejuno/inervação , Jejuno/microbiologia , Lacticaseibacillus rhamnosus/fisiologia , Condução Nervosa , Probióticos , Nervo Vago/fisiologia , Potenciais de Ação , Vias Aferentes/fisiologia , Animais , Sistema Nervoso Entérico/cirurgia , Masculino , Mecanotransdução Celular , Camundongos , Pressão , Tempo de Reação , Fatores de Tempo , Vagotomia , Nervo Vago/cirurgia
6.
FASEB J ; 24(10): 4078-88, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20519636

RESUMO

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.


Assuntos
Motilidade Gastrointestinal , Jejuno/fisiologia , Lactobacillus , Probióticos , Animais , Masculino , Camundongos , Camundongos Endogâmicos BALB C
7.
Sci Rep ; 11(1): 21130, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34702901

RESUMO

The vagus nerve relays mood-altering signals originating in the gut lumen to the brain. In mice, an intact vagus is required to mediate the behavioural effects of both intraluminally applied selective serotonin reuptake inhibitors and a strain of Lactobacillus with antidepressant-like activity. Similarly, the prodepressant effect of lipopolysaccharide is vagus nerve dependent. Single vagal fibres are broadly tuned to respond by excitation to both anti- and prodepressant agents, but it remains unclear how neural responses encode behaviour-specific information. Here we demonstrate using ex vivo experiments that for single vagal fibres within the mesenteric neurovascular bundle supplying the mouse small intestine, a unique neural firing pattern code is common to both chemical and bacterial vagus-dependent antidepressant luminal stimuli. This code is qualitatively and statistically discernible from that evoked by lipopolysaccharide, a non-vagus-dependent antidepressant or control non-antidepressant Lactobacillus strain and are not affected by sex status. We found that all vagus dependent antidepressants evoked a decrease in mean spike interval, increase in spike burst duration, decrease in gap duration between bursts and increase in intra-burst spike intervals. Our results offer a novel neuronal electrical perspective as one explanation for mechanisms of action of gut-derived vagal dependent antidepressants. We expect that our ex vivo individual vagal fibre recording model will improve the design and operation of new, extant electroceutical vagal stimulation devices currently used to treat major depression. Furthermore, use of this vagal antidepressant code should provide a valuable screening tool for novel potential oral antidepressant candidates in preclinical animal models.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Antidepressivos , Lactobacillus/química , Inibidores Seletivos de Recaptação de Serotonina , Nervo Vago/fisiopatologia , Animais , Antidepressivos/química , Antidepressivos/farmacologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Inibidores Seletivos de Recaptação de Serotonina/química , Inibidores Seletivos de Recaptação de Serotonina/farmacologia
8.
PLoS One ; 15(1): e0225481, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31910436

RESUMO

Microvesicles are small lipid, bilayer structures (20-400 nm in diameter) secreted by bacteria, fungi, archaea and parasites involved in inter-bacterial communication and host-pathogen interactions. Lactobacillus reuteri DSM-17938 (DSM) has been shown to have clinical efficacy in the treatment of infantile colic, diarrhea and constipation. We have shown previously that luminal administration to the mouse gut promotes reduction of jejunal motility but increases that in the colon. The production of microvesicles by DSM has been characterized, but the effect of these microvesicles on gastrointestinal motility has yet to be evaluated. To investigate a potential mechanism for the effects of DSM on the intestine, the bacteria and its products have here been tested for changes in velocity, frequency, and amplitude of contractions in intact segments of jejunum and colon excised from mice. The effect of the parent bacteria (DSM) was compared to the conditioned media in which it was grown, and the microvesicles it produced. The media used to culture the bacteria (broth) was tested as a negative control and the conditioned medium was tested after the microvesicles had been removed. DSM, conditioned medium, and the microvesicles all produced comparable effects in both the jejunum and the colon. The treatments individually decreased the velocity and frequency of propagating contractile cluster contractions in the jejunum and increased them in the colon to a similar degree. The broth control had little effect in both tissues. Removal of the microvesicles from the conditioned medium almost completely eradicated their effect on motility in both tissues. These results show that the microvesicles from DSM alone can completely reproduce the effects of the whole bacteria on gut motility. Furthermore, they suggest a new approach to the formulation of orally active bacterial therapeutics and offer a novel way to begin to identify the active bacterial components.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Limosilactobacillus reuteri/metabolismo , Probióticos/metabolismo , Animais , Cólica/metabolismo , Cólica/microbiologia , Colo/microbiologia , Constipação Intestinal/metabolismo , Constipação Intestinal/microbiologia , Diarreia/metabolismo , Diarreia/microbiologia , Motilidade Gastrointestinal/genética , Humanos , Jejuno/metabolismo , Jejuno/microbiologia , Camundongos
9.
Sci Rep ; 10(1): 12978, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32737381

RESUMO

The gut-microbiota-brain axis is implicated in the development of behavioural disorders in mammals. As such, its potential role in disruptive feather pecking (FP) in birds cannot be ignored. Birds with a higher propensity to perform FP have distinct microbiota profiles and feed transit times compared to non-pecking counterparts. Consequently, we hypothesize that the gut microbiota is intimately linked to FP and gut motility, which presents the possibility of using probiotics to control FP behaviour. In the present study, we aim to assess the relationship between cecal motility and the probiotic Lactobacillus rhamnosus in chickens classified as peckers (P, 13 birds) and non-peckers (NP, 17 birds). We show that cecal contractions were 68% less frequent and their amplitude increased by 58% in the presence of L. rhamnosus. Furthermore, the number of FP bouts performed by P birds was positively correlated with contraction velocity and amplitude. We present the first account of gut motility measurements in birds with distinct FP phenotypes. Importantly, the present work demonstrates the clear impact of a probiotic on cecal contractions. These findings lay the foundation for identifying biological differences between P and NP birds which will support the development of FP control strategies.


Assuntos
Ceco , Galinhas/fisiologia , Plumas , Microbioma Gastrointestinal/fisiologia , Motilidade Gastrointestinal , Lacticaseibacillus rhamnosus , Probióticos/farmacologia , Animais , Ceco/microbiologia , Ceco/fisiologia , Feminino , Motilidade Gastrointestinal/efeitos dos fármacos , Motilidade Gastrointestinal/fisiologia
10.
J Parkinsons Dis ; 10(4): 1477-1491, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32925094

RESUMO

BACKGROUND: Parkinson's disease (PD) is a progressive neurodegenerative disorder thought to be caused by accumulation of α-synuclein (α-syn) within the brain, autonomic nerves, and the enteric nervous system (ENS). Involvement of the ENS in PD often precedes the onset of the classic motor signs of PD by many years at a time when severe constipation represents a major morbidity. Studies conducted in vitro and in vivo, have shown that squalamine, a zwitterionic amphipathic aminosterol, originally isolated from the liver of the dogfish shark, effectively displaces membrane-bound α-syn. OBJECTIVE: Here we explore the electrophysiological effect of squalamine on the gastrointestinal (GI) tract of mouse models of PD engineered to express the highly aggregating A53T human α-syn mutant. METHODS: GI motility and in vivo response to oral squalamine in PD model mice and controls were assessed using an in vitro tissue motility protocol and via fecal pellet output. Vagal afferent response to squalamine was measured using extracellular mesenteric nerve recordings from the jejunum. Whole cell patch clamp was performed to measure response to squalamine in the myenteric plexus. RESULTS: Squalamine effectively restores disordered colonic motility in vivo and within minutes of local application to the bowel. We show that topical squalamine exposure to intrinsic primary afferent neurons (IPANs) of the ENS rapidly restores excitability. CONCLUSION: These observations may help to explain how squalamine may promote gut propulsive activity through local effects on IPANs in the ENS, and further support its possible utility in the treatment of constipation in patients with PD.


Assuntos
Constipação Intestinal/tratamento farmacológico , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Sistema Nervoso Entérico/efeitos dos fármacos , Motilidade Gastrointestinal/efeitos dos fármacos , Plexo Mientérico/efeitos dos fármacos , Neurônios Aferentes/efeitos dos fármacos , Doença de Parkinson/complicações , Nervo Vago/efeitos dos fármacos , Animais , Colestanóis/administração & dosagem , Colestanóis/farmacologia , Constipação Intestinal/etiologia , Modelos Animais de Doenças , Jejuno/inervação , Camundongos , Camundongos Transgênicos , Proteínas Mutantes , Neurônios Aferentes/citologia , Técnicas de Patch-Clamp , alfa-Sinucleína/metabolismo
11.
J Cell Mol Med ; 13(8B): 2261-2270, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19210574

RESUMO

Probiotics are live non-pathogenic commensal organisms that exert therapeutic effects in travellers' diarrhea, irritable bowel syndrome and inflammatory bowel disease. Little is known about mechanisms of action of commensal bacteria on intestinal motility and motility-induced pain. It has been proposed that probiotics affect intestinal nerve function, but direct evidence for this has thus far been lacking. We hypothesized that probiotic effects might be mediated by actions on colonic intrinsic sensory neurons. We first determined whether sensory neurons were present in rat colon by their responses to chemical mucosal stimulation and identified them in terms of physiological phenotype and soma morphotype. Enteric neuron excitability and ion channel activity were measured using patch clamp recordings. We fed 10(9)Lactobacillus reuteri (LR) or vehicle control to rats for 9 days. LR ingestion increased excitability (threshold for evoking action potentials) and number of action potentials per depolarizing pulse, decreased calcium-dependent potassium channel (IK(Ca)) opening and decreased the slow afterhyperpolarization (sAHP) in sensory AH neurons, similar to the IK(Ca) antagonists Tram-34 and clotrimazole. LR did not affect threshold for action potential generation in S neurons. Our results demonstrate that LR targets an ion channel in enteric sensory nerves through which LR may affect gut motility and pain perception.


Assuntos
Colo/fisiologia , Ativação do Canal Iônico/fisiologia , Limosilactobacillus reuteri/fisiologia , Neurônios/fisiologia , Canais de Potássio Cálcio-Ativados/antagonistas & inibidores , Canais de Potássio/fisiologia , Animais , Colo/citologia , Masculino , Canais de Potássio Cálcio-Ativados/fisiologia , Ratos , Ratos Sprague-Dawley
12.
Front Neurosci ; 13: 955, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31551703

RESUMO

There is a general decline in gastrointestinal function in old age including decreased intestinal motility, sensory signaling, and afferent sensitivity. There is also increased prevalence of significant constipation in aged populations. We hypothesized this may be linked to reduced colonic motility and alterations in vagal-gut-brain sensory signaling. Using in vitro preparations from young (3 months) and old (18-24 months) male CD1 mice we report functional age-related differences in colonic motility and jejunal mesenteric afferent firing. Furthermore, we tested the effect of the aminosterol squalamine on colonic motility and jejunal vagal firing rate. Old mice had significantly reduced velocity of colonic migrating motor complexes (MMC) by 27% compared to young mice (p = 0.0161). Intraluminal squalamine increased colonic MMC velocity by 31% in old mice (p = 0.0150), which also had significantly reduced mesenteric afferent single-unit firing rates from the jejunum by 51% (p < 0.0001). The jejunal vagal afferent firing rate was reduced in aged mice by 62% (p = 0.0004). While the time to peak response to squalamine was longer in old mice compared to young mice (18.82 ± 1.37 min vs. 12.95 ± 0.99 min; p = 0.0182), it significantly increased vagal afferent firing rate by 36 and 56% in young and old mice, respectively (p = 0.0006, p = 0.0013). Our results show for the first time that the jejunal vagal afferent firing rate is reduced in aged-mice. They also suggest that there is translational potential for the therapeutic use of squalamine in the treatment of age-related constipation and dysmotility.

14.
Nat Commun ; 4: 1465, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23403566

RESUMO

Symbionts or probiotics are known to affect the nervous system. To understand the mechanisms involved, it is important to measure sensory neuron responses and identify molecules responsible for this interaction. Here we test the effects of adding Lactobacillus rhamnosus (JB-1) and Bacteroides fragilis to the epithelium while making voltage recordings from intestinal primary afferent neurons. Sensory responses are recorded within 8 s of applying JB-1 and excitability facilitated within 15 min. Bacteroides fragilis produces similar results, as does its isolated, capsular exopolysaccharide, polysaccharide A. Lipopolysaccharide-free polysaccharide A completely mimics the neuronal effects of the parent organism. Experiments with a mutant Bacteroides fragilis devoid of polysaccharide A shows that polysaccharide A is necessary and sufficient for the neuronal effects. Complex carbohydrates have not been reported before as candidates for such signalling between symbionts and the host. These observations indicate new neuronal targets and invite further study of bacterial carbohydrates as inter-kingdom signalling molecules between beneficial bacteria and sensory neurons.


Assuntos
Bacteroides fragilis/metabolismo , Intestinos/inervação , Intestinos/fisiologia , Polissacarídeos Bacterianos/farmacologia , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Dissecação , Epitélio/efeitos dos fármacos , Epitélio/fisiologia , Técnicas In Vitro , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Lacticaseibacillus rhamnosus/metabolismo , Camundongos , Técnicas de Patch-Clamp , Pirazóis/farmacologia
15.
Clin Vaccine Immunol ; 20(6): 818-26, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23536695

RESUMO

Enteric parasite infections around the world are a huge economic burden and decrease the quality of life for many people. The use of beneficial bacteria has attracted attention for their potential therapeutic applications in various diseases. However, the effects of beneficial bacteria in enteric parasitic infections remain largely unexplored. We investigated the effects of ingestion of Lactobacillus rhamnosus (JB-1) in a model of enteric nematode (Trichuris muris) infection. C57BL/6 (resistant to infection), AKR (susceptible to infection), interleukin 10 (IL-10) knockout (KO), and mucin Muc2 KO mice were infected with T. muris and treated orally with probiotic JB-1 or medium. The mice were sacrificed on various days postinfection to examine goblet cells, epithelial cell proliferation, cytokines, and worm burdens. Treatment with JB-1 significantly enhanced worm expulsion in resistant C57BL/6 mice, and this was associated with increases in IL-10 levels, goblet cell numbers, and epithelial cell proliferation. Beneficial effects of JB-1 were absent in IL-10 KO and resistant mice treated with γ-irradiated bacteria. Live JB-1 treatment also expedited worm expulsion in Muc2 KO mice and, more importantly, in AKR mice (susceptible to infection). Injection of IL-10 directly into the colonic tissue of uninfected mice induced goblet cell hyperplasia. These findings demonstrate that JB-1 modulates goblet cell biology and promotes parasite expulsion via an IL-10-mediated pathway and provide novel insights into probiotic effects on innate defense in nematode infection.


Assuntos
Dieta/métodos , Imunidade Inata , Enteropatias Parasitárias/imunologia , Lacticaseibacillus rhamnosus/imunologia , Tricuríase/imunologia , Animais , Modelos Animais de Doenças , Interleucina-10/imunologia , Enteropatias Parasitárias/parasitologia , Enteropatias Parasitárias/patologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tricuríase/parasitologia , Tricuríase/patologia , Trichuris/imunologia
16.
Am J Physiol Gastrointest Liver Physiol ; 296(4): G868-75, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19179624

RESUMO

Lactobacillus species ingestion can decrease autonomic responses and spinal fiber discharge to nociceptive colorectal distension (CRD), even in the absence of inflammation. The present study aimed to determine whether dorsal root ganglion (DRG) somas could be a locus where the antinociceptive probiotic may have an effect. Healthy rats were fed with Lactobacillus reuteri or vehicle control for 9 days whereupon they were anesthetized, and intermittent distal colonic CRD at 80 mmHg distension was either performed for 1 h or not. The animals were immediately euthanized and patch-clamp recordings taken after isolation and overnight culture from those DRG that projected to the distal colon. CRD decreased the threshold for action potential generation and increased the number of spikes discharged during a standard depolarizing test stimulus, and this effect was blocked by prior probiotic ingestion. The increase in excitability was paralleled by an increase in DRG capacitance, which was not altered by Lactobacillus reuteri ingestion. CRD did not increase tissue weight or myeloperoxidase activity. We suggest that the effects of CRD may have been caused by activity-dependent neurotransmission between DRG somas. CRD evoked increases in action potential upstroke speed, which suggests that it may also have led to augmentation of sodium channel conductances. Probiotic ingestion may have interfered with this hypothetical mechanism since it blocked the effect of CRD on the action potential.


Assuntos
Colo/inervação , Gânglios Espinais/citologia , Limosilactobacillus reuteri , Neurônios/fisiologia , Dor/prevenção & controle , Probióticos/farmacologia , Animais , Eletrofisiologia , Inflamação , Masculino , Neurônios/citologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA