Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Mol Gastroenterol Hepatol ; 10(2): 225-244, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32289500

RESUMO

BACKGROUND & AIMS: Despite achieving endoscopic remission, more than 20% of inflammatory bowel disease patients experience chronic abdominal pain. These patients have increased rectal transient receptor potential vanilloid-1 receptor (TRPV1) expression, a key transducer of inflammatory pain. Because inflammatory bowel disease patients in remission exhibit dysbiosis and microbial manipulation alters TRPV1 function, our goal was to examine whether microbial perturbation modulated transient receptor potential function in a mouse model. METHODS: Mice were given dextran sodium sulfate (DSS) to induce colitis and were allowed to recover. The microbiome was perturbed by using antibiotics as well as fecal microbial transplant (FMT). Visceral and somatic sensitivity were assessed by recording visceromotor responses to colorectal distention and using hot plate/automated Von Frey tests, respectively. Calcium imaging of isolated dorsal root ganglia neurons was used as an in vitro correlate of nociception. The microbiome composition was evaluated via 16S rRNA gene variable region V4 amplicon sequencing, whereas fecal short-chain fatty acids (SCFAs) were assessed by using targeted mass spectrometry. RESULTS: Postinflammatory DSS mice developed visceral and somatic hyperalgesia. Antibiotic administration during DSS recovery induced visceral, but not somatic, hyperalgesia independent of inflammation. FMT of postinflammatory DSS stool into antibiotic-treated mice increased visceral hypersensitivity, whereas FMT of control stool reversed antibiotics' sensitizing effects. Postinflammatory mice exhibited both increased SCFA-producing species and fecal acetate/butyrate content compared with controls. Capsaicin-evoked calcium responses were increased in naive dorsal root ganglion neurons incubated with both sodium butyrate/propionate alone and with colonic supernatants derived from postinflammatory mice. CONCLUSIONS: The microbiome plays a central role in postinflammatory visceral hypersensitivity. Microbial-derived SCFAs can sensitize nociceptive neurons and may contribute to the pathogenesis of postinflammatory visceral pain.


Assuntos
Colite Ulcerativa/complicações , Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Dor Visceral/imunologia , Animais , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/imunologia , Colite Ulcerativa/microbiologia , Colo/efeitos dos fármacos , Colo/imunologia , Colo/microbiologia , Colo/patologia , Sulfato de Dextrana/administração & dosagem , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Disbiose/microbiologia , Ácidos Graxos Voláteis/análise , Ácidos Graxos Voláteis/metabolismo , Fezes/química , Fezes/microbiologia , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Masculino , Camundongos , Nociceptividade , Nociceptores/imunologia , Nociceptores/metabolismo , Canais de Cátion TRPV/metabolismo , Dor Visceral/microbiologia
2.
Biochim Biophys Acta Mol Basis Dis ; 1866(1): 165534, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31634534

RESUMO

Visceral pain, characterized by abdominal discomfort, originates from organs in the abdominal cavity and is a characteristic symptom in patients suffering from irritable bowel syndrome, vulvodynia or interstitial cystitis. Most organs in which visceral pain originates are in contact with the external milieu and continuously exposed to microbes. In order to maintain homeostasis and prevent infections, the immune- and nervous system in these organs cooperate to sense and eliminate (harmful) microbes. Recognition of microbial components or products by receptors expressed on cells from the immune and nervous system can activate immune responses but may also cause pain. We review the microbial compounds and their receptors that could be involved in visceral pain development.


Assuntos
Microbiota/imunologia , Dor Visceral/imunologia , Dor Visceral/microbiologia , Animais , Humanos , Imunidade/imunologia , Sistema Nervoso/microbiologia , Dor Visceral/etiologia
3.
Br J Anaesth ; 123(5): 637-654, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31551115

RESUMO

The relationship between gut microbiota and neurological diseases, including chronic pain, has received increasing attention. The gut microbiome is a crucial modulator of visceral pain, whereas recent evidence suggests that gut microbiota may also play a critical role in many other types of chronic pain, including inflammatory pain, headache, neuropathic pain, and opioid tolerance. We present a narrative review of the current understanding on the role of gut microbiota in pain regulation and discuss the possibility of targeting gut microbiota for the management of chronic pain. Numerous signalling molecules derived from gut microbiota, such as by-products of microbiota, metabolites, neurotransmitters, and neuromodulators, act on their receptors and remarkably regulate the peripheral and central sensitisation, which in turn mediate the development of chronic pain. Gut microbiota-derived mediators serve as critical modulators for the induction of peripheral sensitisation, directly or indirectly regulating the excitability of primary nociceptive neurones. In the central nervous system, gut microbiota-derived mediators may regulate neuroinflammation, which involves the activation of cells in the blood-brain barrier, microglia, and infiltrating immune cells, to modulate induction and maintenance of central sensitisation. Thus, we propose that gut microbiota regulates pain in the peripheral and central nervous system, and targeting gut microbiota by diet and pharmabiotic intervention may represent a new therapeutic strategy for the management of chronic pain.


Assuntos
Dor Crônica/microbiologia , Microbioma Gastrointestinal/fisiologia , Manejo da Dor/métodos , Barreira Hematoencefálica/fisiopatologia , Dor Crônica/fisiopatologia , Dor Crônica/terapia , Trato Gastrointestinal/inervação , Trato Gastrointestinal/microbiologia , Humanos , Inflamação/microbiologia , Neuroimunomodulação/fisiologia , Dor Visceral/microbiologia , Dor Visceral/terapia
4.
Am J Physiol Gastrointest Liver Physiol ; 317(3): G363-G372, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31290688

RESUMO

The potential role of the intestinal microbiota in modulating visceral pain has received increasing attention during recent years. This has led to the identification of signaling pathways that have been implicated in communication between gut bacteria and peripheral pain pathways. In addition to the well-characterized impact of the microbiota on the immune system, which in turn affects nociceptor excitability, bacteria can modulate visceral afferent pathways by effects on enterocytes, enteroendocrine cells, and the neurons themselves. Proteases produced by bacteria, or by host cells in response to bacteria, can increase or decrease the excitability of nociceptive dorsal root ganglion (DRG) neurons depending on the receptor activated. Short-chain fatty acids generated by colonic bacteria are involved in gut-brain communication, and intracolonic short-chain fatty acids have pronociceptive effects in rodents but may be antinociceptive in humans. Gut bacteria modulate the synthesis and release of enteroendocrine cell mediators, including serotonin and glucagon-like peptide-1, which activate extrinsic afferent neurons. Deciphering the complex interactions between visceral afferent neurons and the gut microbiota may lead to the development of improved probiotic therapies for visceral pain.


Assuntos
Colo/microbiologia , Gânglios Espinais/microbiologia , Microbioma Gastrointestinal/fisiologia , Microbiota , Dor Visceral/microbiologia , Animais , Colo/fisiologia , Gânglios Espinais/fisiologia , Humanos , Microbiota/fisiologia , Neurônios Aferentes/microbiologia , Dor Visceral/terapia
5.
J Gastroenterol Hepatol ; 34(8): 1368-1376, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30402954

RESUMO

BACKGROUND AND AIM: Emerging evidence indicates that psychological stress is involved in the pathogenesis of irritable bowel syndrome, which is characterized by visceral hypersensitivity and may be accompanied by gut dysbiosis. However, how such stress contributes to the development of visceral hypersensitivity is incompletely understood. Here, we aimed to investigate the influence that stress-induced microbial changes exert on visceral sensitivity, as well as the possible underlying mechanisms associated with this effect. METHODS: Male Sprague-Dawley rats underwent chronic water avoidance stress (WAS) to induce visceral hypersensitivity. Visceral sensitivity, colonic tight junction protein expression, and short-chain fatty acids of cecal contents were measured. Fecal samples were collected to characterize microbiota profiles. In a separate study, oral gavage of Roseburia in WAS rats was conducted to verify its potential role in the effectiveness on visceral hypersensitivity. RESULTS: Repeated WAS caused visceral hypersensitivity, altered fecal microbiota composition and function, and decreased occludin expression in the colon. Stressed rats exhibited reduced representation of pathways involved in the metabolism of butyrate and reduced abundance of several operational taxonomic units associated with butyrate-producing bacteria, such as Lachnospiraceae. Consistently, supplementation with Roseburia hominis, a species belonging to Lachnospiraceae, significantly increased cecal butyrate content. Moreover, Roseburia supplementation alleviated visceral hypersensitivity and prevented the decreased expression of occludin. CONCLUSIONS: Reduction in the abundance of butyrate-producing Lachnospiraceae, which is beneficial for the intestinal barrier, was involved in the formation of visceral hypersensitivity. R. hominis is a potential probiotic for treating stress-induced visceral hypersensitivity.


Assuntos
Butiratos/metabolismo , Clostridiales/metabolismo , Colo/microbiologia , Hiperalgesia/prevenção & controle , Limiar da Dor , Probióticos/farmacologia , Estresse Psicológico/complicações , Dor Visceral/prevenção & controle , Animais , Colo/metabolismo , Modelos Animais de Doenças , Fezes/microbiologia , Microbioma Gastrointestinal , Hiperalgesia/etiologia , Hiperalgesia/microbiologia , Hiperalgesia/fisiopatologia , Masculino , Ocludina/metabolismo , Percepção da Dor , Ratos Sprague-Dawley , Junções Íntimas/metabolismo , Junções Íntimas/microbiologia , Dor Visceral/etiologia , Dor Visceral/microbiologia , Dor Visceral/fisiopatologia
6.
Neurogastroenterol Motil ; 30(6): e13299, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29383802

RESUMO

BACKGROUND: Irritable bowel syndrome (IBS) is a common gastrointestinal disorder associated with altered gastrointestinal microflora and increased nociception to colonic distension. This visceral hypersensitivity can be reversed in our rat maternal separation model by fungicides. Menthacarin® is a proprietary combination of essential oils from Mentha x piperita L. and Carum carvi. Because these oils exhibit antifungal and antibacterial properties, we investigated whether Menthacarin® can reverse existing visceral hypersensitivity in maternally separated rats. METHODS: In non-handled and maternally separated rats, we used the visceromotor responses to colorectal distension as measure for visceral sensitivity. We evaluated this response before and 24 hours after water-avoidance stress and after 7 days treatment with Menthacarin® or control. The pre- and post-treatment mycobiome and microbiome were characterized by sequencing of fungal internal transcribed spacer 1 (ITS-1) and bacterial 16s rDNA regions. In vitro antifungal and antimicrobial properties of Menthacarin® were studied with radial diffusion assay. KEY RESULTS: Menthacarin® inhibited in vitro growth of yeast and bacteria. Water-avoidance caused visceral hypersensitivity in maternally separated rats, and this was reversed by treatment. Multivariate analyses of ITS-1 and 16S high throughput data showed that maternal separation, induced changes in the myco- and microbiome. Menthacarin® treatment of non-handled and maternally separated rats shifted the mycobiomes to more similar compositions. CONCLUSIONS & INFERENCES: The development of visceral hypersensitivity in maternally separated rats and the Menthacarin® -mediated reversal of hypersensitivity is associated with changes in the mycobiome. Therefore, Menthacarin® may be a safe and effective treatment option that should be tested for IBS.


Assuntos
Hiperalgesia/tratamento farmacológico , Micobioma/efeitos dos fármacos , Óleos Voláteis/administração & dosagem , Óleos de Plantas/administração & dosagem , Dor Visceral/tratamento farmacológico , Animais , Animais Recém-Nascidos , Antibacterianos/administração & dosagem , Antibacterianos/isolamento & purificação , Antifúngicos/administração & dosagem , Antifúngicos/isolamento & purificação , Combinação de Medicamentos , Hiperalgesia/microbiologia , Hiperalgesia/psicologia , Masculino , Privação Materna , Mentha piperita , Micobioma/fisiologia , Óleos Voláteis/isolamento & purificação , Óleos de Plantas/isolamento & purificação , Ratos , Ratos Long-Evans , Dor Visceral/microbiologia , Dor Visceral/psicologia
7.
Acta Biochim Biophys Sin (Shanghai) ; 50(2): 216-223, 2018 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-29329362

RESUMO

Visceral hypersensitivity induced by stress is quite common in irritable bowel syndrome (IBS) patients. Probiotics play an important role in reducing visceral hypersensitivity in IBS patients. However, the mechanism has not been clearly elucidated. In this study, we investigated the role of nod-like receptor pyrin domain-containing protein 6 (NLRP6) in Clostridium butyricum-regulated IBS induced by stress. Our results showed that NLRP6 was down-regulated in IBS group colon tissues. In addition, IL-18, IL-1ß, myeloperoxidase (MPO), d-lactic acid (D-LA), and CD172a were up-regulated in the IBS group of colonic mucous. IL-18 and IL-1ß were also increased after the NLRP6 gene was silenced. Pathological score suggested low inflammation of colonic mucous rather than terminal ileum. Water-avoidance stress (WAS) showed visceral hypersensitivity to colonic distension. However, treatment with Clostridium butyricum reversed these results, exerting a beneficial effect. In conclusion, Clostridium butyricum may exert a beneficial action on visceral hypersensitivity of IBS by inhibiting low grade inflammation of colonic mucous through its action on NLRP6.


Assuntos
Clostridium butyricum/fisiologia , Inflamação/microbiologia , Mucosa Intestinal/microbiologia , Síndrome do Intestino Irritável/microbiologia , Receptores de Superfície Celular/metabolismo , Dor Visceral/microbiologia , Animais , Colo/metabolismo , Colo/microbiologia , Colo/patologia , Feminino , Interações Hospedeiro-Patógeno , Humanos , Inflamação/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Síndrome do Intestino Irritável/metabolismo , Camundongos Endogâmicos C57BL , Probióticos/administração & dosagem , Interferência de RNA , Receptores de Superfície Celular/genética , Dor Visceral/metabolismo
8.
Gastroenterology ; 153(6): 1594-1606.e2, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28865734

RESUMO

BACKGROUND & AIMS: Separation of newborn rats from their mothers induces visceral hypersensitivity and impaired epithelial secretory cell lineages when they are adults. Little is known about the mechanisms by which maternal separation causes visceral hypersensitivity or its relationship with defects in epithelial secretory cell lineages. METHODS: We performed studies with C3H/HeN mice separated from their mothers as newborns and mice genetically engineered (Sox9flox/flox-vil-cre on C57BL/6 background) to have deficiencies in Paneth cells. Paneth cell deficiency was assessed by lysozyme staining of ileum tissues and lysozyme activity in fecal samples. When mice were 50 days old, their abdominal response to colorectal distension was assessed by electromyography. Fecal samples were collected and microbiota were analyzed using Gut Low-Density Array quantitative polymerase chain reaction. RESULTS: Mice with maternal separation developed visceral hypersensitivity and defects in Paneth cells, as reported from rats, compared with mice without maternal separation. Sox9flox/flox-vil-Cre mice also had increased visceral hypersensitivity compared with control littermate Sox9flox/flox mice. Fecal samples from mice with maternal separation and from Sox9flox/flox-vil-cre mice had evidence for intestinal dysbiosis of the microbiota, characterized by expansion of Escherichia coli. Daily gavage of conventional C3H/HeN adult mice with 109 commensal E coli induced visceral hypersensitivity. Conversely, daily oral administration of lysozyme prevented expansion of E coli during maternal separation and visceral hypersensitivity. CONCLUSIONS: Mice with defects in Paneth cells (induced by maternal separation or genetically engineered) have intestinal expansion of E coli leading to visceral hypersensitivity. These findings provide evidence that Paneth cell function and intestinal dysbiosis are involved in visceral sensitivity.


Assuntos
Ansiedade de Separação/complicações , Escherichia coli/crescimento & desenvolvimento , Microbioma Gastrointestinal , Hiperalgesia/etiologia , Celulas de Paneth/microbiologia , Dor Visceral/etiologia , Fatores Etários , Animais , Animais Recém-Nascidos , Ansiedade de Separação/metabolismo , Ansiedade de Separação/microbiologia , Ansiedade de Separação/fisiopatologia , Modelos Animais de Doenças , Disbiose , Fezes/microbiologia , Feminino , Predisposição Genética para Doença , Hiperalgesia/metabolismo , Hiperalgesia/microbiologia , Hiperalgesia/fisiopatologia , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Muramidase/administração & dosagem , Muramidase/metabolismo , Celulas de Paneth/metabolismo , Fenótipo , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Dor Visceral/metabolismo , Dor Visceral/microbiologia , Dor Visceral/fisiopatologia
9.
Handb Exp Pharmacol ; 239: 269-287, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28035535

RESUMO

A growing body of preclinical and clinical evidence supports a relationship between the complexity and diversity of the microorganisms that inhabit our gut (human gastrointestinal microbiota) and health status. Under normal homeostatic conditions this microbial population helps maintain intestinal peristalsis, mucosal integrity, pH balance, immune priming and protection against invading pathogens. Furthermore, these microbes can influence centrally regulated emotional behaviour through mechanisms including microbially derived bioactive molecules (amino acid metabolites, short-chain fatty acids, neuropeptides and neurotransmitters), mucosal immune and enteroendocrine cell activation, as well as vagal nerve stimulation.The microbiota-gut-brain axis comprises a dynamic matrix of tissues and organs including the brain, autonomic nervous system, glands, gut, immune cells and gastrointestinal microbiota that communicate in a complex multidirectional manner to maintain homeostasis and resist perturbation to the system. Changes to the microbial environment, as a consequence of illness, stress or injury, can lead to a broad spectrum of physiological and behavioural effects locally including a decrease in gut barrier integrity, altered gut motility, inflammatory mediator release as well as nociceptive and distension receptor sensitisation. Centrally mediated events including hypothalamic-pituitary-adrenal (HPA) axis, neuroinflammatory events and neurotransmitter systems are concomitantly altered. Thus, both central and peripheral pathways associated with pain manifestation and perception are altered as a consequence of the microbiota-gut-brain axis imbalance.In this chapter the involvement of the gastrointestinal microbiota in visceral pain is reviewed. We focus on the anatomical and physiological nodes whereby microbiota may be mediating pain response, and address the potential for manipulating gastrointestinal microbiota as a therapeutic target for visceral pain.


Assuntos
Dor Abdominal/microbiologia , Microbioma Gastrointestinal , Trato Gastrointestinal/inervação , Trato Gastrointestinal/microbiologia , Dor Visceral/microbiologia , Dor Visceral/fisiopatologia , Dor Abdominal/fisiopatologia , Dor Abdominal/terapia , Animais , Encéfalo/fisiopatologia , Interações Hospedeiro-Patógeno , Humanos , Vias Neurais/microbiologia , Vias Neurais/fisiopatologia , Percepção da Dor , Limiar da Dor , Probióticos/uso terapêutico , Dor Visceral/terapia
10.
Neuroscience ; 339: 463-477, 2016 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-27742460

RESUMO

Gut microbiota colonization is a key event for host physiology that occurs early in life. Disruption of this process leads to altered brain development which ultimately manifests as changes in brain function and behaviour in adulthood. Studies using germ-free (GF) mice highlight the extreme impact on brain health that results from life without commensal microbes. However, the impact of microbiota disturbances occurring in adulthood is less studied. To this end, we depleted the gut microbiota of 10-week-old male SpragueDawley rats via chronic antibiotic treatment. Following this marked, sustained depletion of the gut bacteria, we investigated behavioural and molecular hallmarks of gut-brain communication. Our results reveal that depletion of the gut microbiota during adulthood results in deficits in spatial memory as tested by Morris water maze, decreased visceral sensitivity and a greater display of depressive-like behaviours in the forced swim test. In tandem with these clear behavioural alterations we found changes in altered CNS serotonin concentration along with changes in the mRNA levels of corticotrophin releasing hormone receptor 1 and glucocorticoid receptor. Additionally, we found changes in the expression of brain derived neurotrophic factor (BDNF), a hallmark of altered microbiota-gut-brain axis signalling. In summary, this model of antibiotic-induced depletion of the gut microbiota can be used for future studies interested in the impact of the gut microbiota on host health without the confounding developmental influence of early-life microbial alterations.


Assuntos
Encéfalo/metabolismo , Depressão/microbiologia , Microbioma Gastrointestinal , Transtornos da Memória/microbiologia , Dor Visceral/microbiologia , Animais , Antibacterianos/farmacologia , Ansiedade/microbiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Ceco/efeitos dos fármacos , Ceco/metabolismo , Corticosterona/sangue , Depressão/metabolismo , Fezes/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Masculino , Transtornos da Memória/metabolismo , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Receptores de Glucocorticoides/metabolismo , Serotonina/metabolismo , Dor Visceral/metabolismo , Dor Visceral/psicologia
11.
Behav Brain Res ; 287: 59-72, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25794930

RESUMO

Increasing evidence suggests that a brain-gut-microbiome axis exists, which has the potential to play a major role in modulating behaviour. However, the role of this axis in cognition remains relatively unexplored. Probiotics, which are commensal bacteria offering potential health benefit, have been shown to decrease anxiety, depression and visceral pain-related behaviours. In this study, we investigate the potential of two Bifidobacteria strains to modulate cognitive processes and visceral pain sensitivity. Adult male BALB/c mice were fed daily for 11 weeks with B. longum 1714, B. breve 1205 or vehicle treatment. Starting at week 4, animals were behaviourally assessed in a battery of tests relevant to different aspects of cognition, as well as locomotor activity and visceral pain. In the object recognition test, B. longum 1714-fed mice discriminated between the two objects faster than all other groups and B. breve 1205-fed mice discriminated faster than vehicle animals. In the Barnes maze, B. longum 1714-treated mice made fewer errors than other groups, suggesting a better learning. In the fear conditioning, B. longum 1714-treated group also showed better learning and memory, yet presenting the same extinction learning profile as controls. None of the treatments affected visceral sensitivity. Altogether, these data suggest that B. longum 1714 had a positive impact on cognition and also that the effects of individual Bifidobacteria strains do not generalise across the species. Clinical validation of the effects of probiotics on cognition is now warranted.


Assuntos
Ansiedade/microbiologia , Cognição/efeitos dos fármacos , Probióticos/administração & dosagem , Estresse Psicológico/microbiologia , Animais , Bifidobacterium , Peso Corporal/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Medo/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Atividade Motora/efeitos dos fármacos , Reconhecimento Psicológico/efeitos dos fármacos , Dor Visceral/microbiologia
12.
World J Gastroenterol ; 20(22): 6832-43, 2014 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-24944474

RESUMO

AIM: To investigate the effect of the probiotic combination Lactibiane Tolerance(®) (LT) on epithelial barrier function in vitro and in vivo. METHODS: The effect of the multispecies probiotic LT was assessed on several models of epithelial barrier function both in vitro (in basal and inflammatory conditions) and in vivo [visceral hypersensitivity induced by chronic stress or by colonic perfusion of a fecal supernatant (FSN) from patients with irritable bowel syndrome (IBS)]. In vitro, we measured the permeability of confluent T84 cell monolayers incubated with or without LT by evaluating the paracellular flux of macromolecules, in basal conditions and after stimulation with lipopolysaccharide (LPS) or with conditioned medium of colonic biopsies from IBS patients (IBS-CM). In vivo, male C57/Bl6 mice received orally NaCl or LT for 15 d and were submitted to water avoidance stress (WAS) before evaluating visceral sensitivity by measuring the myoelectrical activity of the abdominal muscle and the paracellular permeability with (51)Cr-EDTA. Permeability and sensitivity were also measured after colonic instillation of FSN. Tight-junctions were assessed by immunoblotting and TLR-4 expression was evaluated by immunohistochemistry RESULTS: Incubation of T84 cell monolayers with LT in basal conditions had no significant effect on permeability (P > 0.05 vs culture medium). By contrast, addition of LT bacterial bodies (LT) completely prevented the LPS-induced increase in paracellular permeability (P < 0.01 vs LPS 10 ng/mL (LPS 10); P < 0.01 vs LPS 100 ng/mL (LPS 100), P > 0.05 vs culture medium). The effect was dose dependent as addition of 10(9) LT bacterial bodies induced a stronger decrease in absorbance than 10(6) LT (10(9) LT + LPS 10: -20.1% ± 13.4, P < 0.01 vs LPS 10; 10(6) LT + LPS 10: -11.6% ± 6.2, P < 0.01 vs LPS 10; 10(9) LT + LPS 100: -14.4% ± 5.5, P < 0.01 vs LPS 100; 10(6) LT + LPS 100: -11.6% ± 7.3, P < 0.05 vs LPS 100). Moreover, the increase in paracellular permeability induced by culturing T84 cells with conditioned medium of colonic biopsies from IBS patients (IBS-CM) was completely inhibited in the presence of 10(9) LT (P < 0.01 vs IBS-CM). LT also significantly prevented the epithelial disruption induced by intracolonic infusion of fecal supernatant from IBS patients (P < 0.01 vs IBS FSN) or water avoidance stress P < 0.01 vs WAS) in C57/Bl6 mice and increased the expression of occludin in vitro and in vivo, as assessed by immnunoblotting. The WAS-induced effect on visceral sensitivity was prevented by LT treatment since values obtained for all steps of colorectal distension were significantly (P < 0.01) different from the WAS group. Finally, LT down-regulated the response mediated through TLR-4 in vitro (decrease in tumor necrosis factor α secretion in response to LPS: -65.8% for 10(9) LT and -52.5% for 10(6) LT, P < 0.01 vs LPS) and in vivo (inhibition of WAS induced an increase in TLR-4 expression in the LT treated mice colon, P < 0.01 vs WAS). CONCLUSION: The probiotic LT mix prevented the disruption to the epithelial barrier induced by LPS, stress or colonic soluble factors from IBS patients and prevented visceral hypersensitivity.


Assuntos
Colo/microbiologia , Células Epiteliais/microbiologia , Mucosa Intestinal/microbiologia , Síndrome do Intestino Irritável/terapia , Probióticos/uso terapêutico , Animais , Linhagem Celular , Colo/efeitos dos fármacos , Colo/metabolismo , Meios de Cultivo Condicionados/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Fezes/microbiologia , Humanos , Hiperalgesia/metabolismo , Hiperalgesia/microbiologia , Hiperalgesia/prevenção & controle , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Síndrome do Intestino Irritável/metabolismo , Síndrome do Intestino Irritável/microbiologia , Lipopolissacarídeos/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Permeabilidade , Proteínas de Junções Íntimas/metabolismo , Junções Íntimas/metabolismo , Junções Íntimas/microbiologia , Fatores de Tempo , Técnicas de Cultura de Tecidos , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Dor Visceral/metabolismo , Dor Visceral/microbiologia , Dor Visceral/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...