Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
FASEB J ; 33(1): 770-781, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30067379

RESUMO

Early-life adversity is a major risk factor for the development of diseases later in life. Maternal protein restriction (MPR) is associated with morbidities in offspring affecting multiple organs, but its impact on the gastrointestinal (GI) tract remains poorly studied. Using a rat model, we examined the consequences of MPR on GI function and on the enteric nervous system (ENS) in the offspring at postnatal d 35 under basal state and following a water avoidance stress (WAS). Compared with control rats, MPR rats exhibited greater colonic motility, permeability, and corticosteronemia. In contrast to controls, MPR rats presented a blunted functional and corticosteronemic response to WAS. Furthermore, MPR rats showed an increased proportion of choline acetyltransferase-immunoreactive (ChAT-IR) neurons and a reduced level of autophagy in colonic myenteric neurons. In ENS cultures, corticosterone treatment increased the proportion of ChAT-IR neurons and reduced autophagy level in enteric neurons. Inhibition of autophagy in ENS cultures resulted in a higher vulnerability of enteric neurons to a cellular stress. Altogether, this study suggests that MPR induced GI dysfunction and ENS alterations in offspring rats and that MPR-induced increased corticosteronemia might be involved in ENS remodeling and altered responsiveness of the gut to stressors later in life.-Aubert, P., Oleynikova, E., Rizvi, H., Ndjim, M., Le Berre-Scoul, C., Grohard, P. A., Chevalier, J., Segain, J.-P., Le Drean, G., Neunlist, M., Boudin, H. Maternal protein restriction induces gastrointestinal dysfunction and enteric nervous system remodeling in rat offspring.


Assuntos
Proteínas Alimentares/administração & dosagem , Sistema Nervoso Entérico/fisiopatologia , Trato Gastrointestinal/fisiopatologia , Exposição Materna , Animais , Autofagia , Tamanho Corporal , Peso Corporal , Colina O-Acetiltransferase/metabolismo , Colo/fisiopatologia , Corticosterona/sangue , Sistema Nervoso Entérico/enzimologia , Feminino , Absorção Intestinal , Modelos Animais , Neurônios/enzimologia , Neurônios/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Ratos Sprague-Dawley
2.
Brain Behav Immun ; 80: 179-192, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30872090

RESUMO

The accumulation of adverse events in utero and during childhood differentially increases the vulnerability to psychiatric diseases in men and women. Gut microbiota is highly sensitive to the early environment and has been recently hypothesized to affect brain development. However, the impact of early-life adversity on gut microbiota, notably with regards to sex differences, remains to be explored. We examined the effects of multifactorial early-life adversity on behavior and microbiota composition in C3H/HeN mice of both sexes exposed to a combination of maternal immune activation (lipopolysaccharide injection on embryonic day 17, 120 µg/kg, i.p.), maternal separation (3hr per day from postnatal day (PND)2 to PND14) and maternal unpredictable chronic mild stress. At adulthood, offspring exposed to multi-hit early adversity showed sex-specific behavioral phenotypes with males exhibiting deficits in social behavior and females showing increased anxiety in the elevated plus maze and increased compulsive behavior in the marble burying test. Early adversity also differentially regulated gene expression in the medial prefrontal cortex (mPFC) according to sex. Interestingly, several genes such as Arc, Btg2, Fosb, Egr4 or Klf2 were oppositely regulated by early adversity in males versus females. Finally, 16S-based microbiota profiling revealed sex-dependent gut dysbiosis. In males, abundance of taxa belonging to Lachnospiraceae and Porphyromonadaceae families or other unclassified Firmicutes, but also Bacteroides, Lactobacillus and Alloprevotella genera was regulated by early adversity. In females, the effects of early adversity were limited and mainly restricted to Lactobacillus and Mucispirillum genera. Our work reveals marked sex differences in a multifactorial model of early-life adversity, both on emotional behaviors and gut microbiota, suggesting that sex should systematically be considered in preclinical studies both in neurogastroenterology and psychiatric research.


Assuntos
Microbioma Gastrointestinal/fisiologia , Estresse Psicológico/metabolismo , Estresse Psicológico/microbiologia , Animais , Animais Recém-Nascidos , Ansiedade/metabolismo , Comportamento Animal/fisiologia , Encéfalo/metabolismo , Disbiose/metabolismo , Feminino , Masculino , Privação Materna , Camundongos , Camundongos Endogâmicos C3H , Microbiota , Córtex Pré-Frontal/metabolismo , Fatores Sexuais , Comportamento Social
3.
J Physiol ; 595(2): 583-598, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27436013

RESUMO

KEY POINTS: Unlike astrocytes in the brain, the potential role of enteric glial cells (EGCs) in the formation of the enteric neuronal circuit is currently unknown. To examine the role of EGCs in the formation of the neuronal network, we developed a novel neuron-enriched culture model from embryonic rat intestine grown in indirect coculture with EGCs. We found that EGCs shape axonal complexity and synapse density in enteric neurons, through purinergic- and glial cell line-derived neurotrophic factor-dependent pathways. Using a novel and valuable culture model to study enteric neuron-glia interactions, our study identified EGCs as a key cellular actor regulating neuronal network maturation. ABSTRACT: In the nervous system, the formation of neuronal circuitry results from a complex and coordinated action of intrinsic and extrinsic factors. In the CNS, extrinsic mediators derived from astrocytes have been shown to play a key role in neuronal maturation, including dendritic shaping, axon guidance and synaptogenesis. In the enteric nervous system (ENS), the potential role of enteric glial cells (EGCs) in the maturation of developing enteric neuronal circuit is currently unknown. A major obstacle in addressing this question is the difficulty in obtaining a valuable experimental model in which enteric neurons could be isolated and maintained without EGCs. We adapted a cell culture method previously developed for CNS neurons to establish a neuron-enriched primary culture from embryonic rat intestine which was cultured in indirect coculture with EGCs. We demonstrated that enteric neurons grown in such conditions showed several structural, phenotypic and functional hallmarks of proper development and maturation. However, when neurons were grown without EGCs, the complexity of the axonal arbour and the density of synapses were markedly reduced, suggesting that glial-derived factors contribute strongly to the formation of the neuronal circuitry. We found that these effects played by EGCs were mediated in part through purinergic P2Y1 receptor- and glial cell line-derived neurotrophic factor-dependent pathways. Using a novel and valuable culture model to study enteric neuron-glia interactions, our study identified EGCs as a key cellular actor required for neuronal network maturation.


Assuntos
Intestinos/embriologia , Neurogênese/fisiologia , Neuroglia/fisiologia , Neurônios/fisiologia , Animais , Células Cultivadas , Técnicas de Cocultura , Embrião de Mamíferos , Feminino , Intestinos/citologia , Gravidez , Ratos Sprague-Dawley
4.
J Physiol ; 594(15): 4309-23, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-26939757

RESUMO

KEY POINTS: Reducing intestinal epithelial barrier (IEB) dysfunctions is recognized as being of major therapeutic interest for various intestinal disorders. Sacral nerve stimulation (SNS) is known to reduce IEB permeability. Here, we report in a pig model that SNS enhances morphological and functional recovery of IEB following mucosal injury induced via 2,4,6-trinitrobenzenesulfonic acid. These effects are associated with an increased expression of tight junction proteins such as ZO-1 and FAK. These results establish that SNS enhances intestinal barrier repair in acute mucosal injury. They further set the scientific basis for future use of SNS as a complementary or alternative therapeutic option for the treatment of gut disorders with IEB dysfunctions such as inflammatory bowel diseases or irritable bowel syndrome. ABSTRACT: Intestinal epithelial barrier (IEB) dysfunctions, such as increased permeability or altered healing, are central to intestinal disorders. Sacral nerve stimulation (SNS) is known to reduce IEB permeability, but its ability to modulate IEB repair remains unknown. This study aimed to characterize the impact of SNS on mucosal repair following 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced lesions. Six pigs were stimulated by SNS 3 h prior to and 3 h after TNBS enema, while sham animals (n = 8) were not stimulated. The impact of SNS on mucosal changes was evaluated by combining in vivo imaging, histological and functional methods. Biochemical and transcriptomic approaches were used to analyse the IEB and mucosal inflammatory response. We observed that SNS enhanced the recovery from TNBS-induced increase in transcellular permeability. At 24 h, TNBS-induced alterations of mucosal morphology were significantly less in SNS compared with sham animals. SNS reduced TNBS-induced changes in ZO-1 expression and its epithelial pericellular distribution, and also increased pFAK/FAK expression compared with sham. Interestingly, SNS increased the mucosal density of neutrophils, which was correlated with an increase in trypsin and TGF-ß1 levels compared with sham. Finally, SNS prevented the TNBS-induced increases in IL-1ß and IL-4 over time that were observed with sham treatment. In conclusion, our results show that SNS enhances mucosal repair following injury. This study highlights novel mechanisms of action of SNS and identifies SNS as a new therapy for diseases with IEB repair disorders.


Assuntos
Mucosa Intestinal/fisiologia , Sacro/inervação , Cicatrização , Animais , Citocinas/sangue , Estimulação Elétrica , Terapia por Estimulação Elétrica , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Reto/inervação , Suínos , Ácido Trinitrobenzenossulfônico
5.
Am J Physiol Gastrointest Liver Physiol ; 310(11): G941-51, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27056724

RESUMO

The postnatal period is crucial for the development of gastrointestinal (GI) functions. The enteric nervous system is a key regulator of GI functions, and increasing evidences indicate that 1) postnatal maturation of enteric neurons affect the development of GI functions, and 2) microbiota-derived short-chain fatty acids can be involved in this maturation. Although enteric glial cells (EGC) are central regulators of GI functions, the postnatal evolution of their phenotype remains poorly defined. We thus characterized the postnatal evolution of EGC phenotype in the colon of rat pups and studied the effect of short-chain fatty acids on their maturation. We showed an increased expression of the glial markers GFAP and S100ß during the first postnatal week. As demonstrated by immunohistochemistry, a structured myenteric glial network was observed at 36 days in the rat colons. Butyrate inhibited EGC proliferation in vivo and in vitro but had no effect on glial marker expression. These results indicate that the EGC myenteric network continues to develop after birth, and luminal factors such as butyrate endogenously produced in the colon may affect this development.


Assuntos
Butiratos/farmacologia , Plexo Mientérico/citologia , Neurogênese , Neuroglia/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Colo/citologia , Colo/crescimento & desenvolvimento , Colo/inervação , Colo/metabolismo , Ácidos Graxos/metabolismo , Feminino , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Plexo Mientérico/crescimento & desenvolvimento , Plexo Mientérico/metabolismo , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fenótipo , Ratos , Ratos Sprague-Dawley , Proteínas S100/genética , Proteínas S100/metabolismo
6.
Am J Physiol Gastrointest Liver Physiol ; 307(3): G274-85, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24875097

RESUMO

The glucagon-like peptide 2 (GLP-2) is an intestinotrophic hormone with growth promoting and anti-inflammatory actions. However, the full biological functions of GLP-2 and the localization of its receptor (GLP-2R) remain controversial. Among cell lines tested, the expression of GLP-2R transcript was detected in human colonic myofibroblasts (CCD-18Co) and in primary culture of rat enteric nervous system but not in intestinal epithelial cell lines, lymphocytes, monocytes, or endothelial cells. Surprisingly, GLP-2R was expressed in murine (GLUTag), but not human (NCI-H716) enteroendocrine cells. The screening of GLP-2R mRNA in mice organs revealed an increasing gradient of GLP-2R toward the distal gut. An unexpected expression was detected in the mesenteric fat, mesenteric lymph nodes, bladder, spleen, and liver, particularly in hepatocytes. In two mice models of trinitrobenzene sulfonic acid (TNBS)- and dextran sulfate sodium (DSS)-induced colitis, the colonic expression of GLP-2R mRNA was decreased by 60% compared with control mice. Also, GLP-2R mRNA was significantly downregulated in intestinal tissues of inflammatory bowel disease patients. Therapeutically, GLP-2 showed a weak restorative effect on intestinal inflammation during TNBS-induced colitis as assessed by macroscopic score and inflammatory markers. Finally, GLP-2 treatment accelerated mouse liver regeneration following partial hepatectomy as assessed by histological and molecular analyses. In conclusion, the limited therapeutic effect of GLP-2 on colonic inflammation dampens its utility in the management of severe inflammatory intestinal disorders. However, the role of GLP-2 in liver regeneration is a novelty that might introduce GLP-2 into the management of liver diseases and emphasizes on the importance of elucidating other extraintestinal functions of GLP-2.


Assuntos
Anti-Inflamatórios/farmacologia , Colite/tratamento farmacológico , Colo/efeitos dos fármacos , Fármacos Gastrointestinais/farmacologia , Peptídeo 2 Semelhante ao Glucagon/farmacologia , Regeneração Hepática/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptores de Glucagon/agonistas , Animais , Células CACO-2 , Colite/induzido quimicamente , Colite/genética , Colite/metabolismo , Colite/patologia , Colo/metabolismo , Colo/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Células Enteroendócrinas/efeitos dos fármacos , Células Enteroendócrinas/metabolismo , Regulação da Expressão Gênica , Receptor do Peptídeo Semelhante ao Glucagon 2 , Células HT29 , Células Hep G2 , Hepatectomia , Humanos , Células Jurkat , Fígado/metabolismo , Fígado/cirurgia , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo , Ratos , Receptores de Glucagon/genética , Receptores de Glucagon/metabolismo , Proteínas Recombinantes/farmacologia , Fatores de Tempo , Ácido Trinitrobenzenossulfônico
7.
J Neuroinflammation ; 11: 202, 2014 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-25497784

RESUMO

BACKGROUND: Evidence continues to mount concerning the importance of the enteric nervous system (ENS) in controlling numerous intestinal functions in addition to motility and epithelial functions. Nevertheless, little is known concerning the direct participation of the ENS in the inflammatory response of the gut during infectious or inflammatory insults. In the present study we analyzed the ENS response to bacterial lipopolysaccharide, in particular the production of a major proinflammatory cytokine, tumor necrosis factor-alpha (TNF-α). METHODS: TNF-α expression (measured by qPCR, quantitative Polymerase Chain Reaction) and production (measured by ELISA) were measured in human longitudinal muscle-myenteric plexus (LMMP) and rat ENS primary cultures (rENSpc). They were either treated or not treated with lipopolysaccharide (LPS) in the presence or not of electrical field stimulation (EFS). Activation of extracellular signal-regulated kinase (ERK) and 5'-adenosine monophosphate-activated protein kinase (AMPK) pathways was analyzed by immunocytochemistry and Western blot analysis. Their implications were studied using specific inhibitors (U0126, mitogen-activated protein kinase kinase, MEK, inhibitor and C compound, AMPK inhibitor). We also analyzed toll-like receptor 2 (TLR2) expression and interleukin-6 (IL-6) production after LPS treatment simultaneously with EFS or TNF-α-neutralizing antibody. RESULTS: Treatment of human LMMP or rENSpc with LPS induced an increase in TNF-α production. Activation of the ENS by EFS significantly inhibited TNF-α production. This regulation occurred at the transcriptional level. Signaling analyses showed that LPS induced activation of ERK but not AMPK, which was constitutively activated in rENSpc neurons. Both U0126 and C compound almost completely prevented LPS-induced TNF-α production. In the presence of LPS, EFS inhibited the ERK and AMPK pathways. In addition, we demonstrated using TNF-α-neutralizing antibody that LPS-induced TNF-α production increased TLR2 expression and reduced IL-6 production. CONCLUSIONS: Our results show that LPS induced TNF-α production by enteric neurons through activation of the canonical ERK pathway and also in an AMPK-dependent manner. ENS activation through the inhibition of these pathways decreased TNF-α production, thereby modulating the inflammatory response induced by endotoxin.


Assuntos
Sistema Nervoso Entérico/metabolismo , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/toxicidade , Neurônios/metabolismo , Animais , Células Cultivadas , Sistema Nervoso Entérico/efeitos dos fármacos , Humanos , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/biossíntese
8.
J Nutr Biochem ; 99: 108865, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34582967

RESUMO

Pain-related functional gastrointestinal disorders (FGIDs) are characterized by visceral hypersensitivity (VHS) associated with alterations in the microbiota-gut-brain axis. Since human milk oligosaccharides (HMOs) modulate microbiota, gut and brain, we investigated whether HMOs impact VHS, and explored the role of gut microbiota. To induce VHS, C57BL/6JRj mice received hourly water avoidance stress (WAS) sessions for 10 d, or antibiotics (ATB) for 12 d. Challenged and unchallenged (Sham) animals were fed AIN93M diet (Cont) or AIN93M containing 1% of a 6-HMO mix (HMO6). VHS was assessed by monitoring the visceromotor response to colorectal distension. Fecal microbiome was analyzed by shotgun metagenomics. The effect of HMO6 sub-blends on VHS and nociceptive pathways was further tested using the WAS model. In mice fed Cont, WAS and ATB increased the visceromotor response to distension. HMO6 decreased WAS-mediated electromyographic rise at most distension volumes and overall Area Under Curve (AUC=6.12±0.50 in WAS/HMO6 vs. 9.46±0.50 in WAS/Cont; P<.0001). In contrast, VHS in ATB animals was not improved by HMO6. In WAS, HMO6 promoted most microbiota taxa and several functional pathways associated with low VHS and decreased those associated with high VHS. Among the sub-blends, 2'FL+DFL and LNT+6'SL reduced visceromotor response close to Sham/Cont values and modulated serotoninergic and CGRPα-related pathways. This research further substantiates the capacity of HMOs to modulate the microbiota-gut-brain communication and identifies mitigation of abdominal pain as a new HMO benefit. Ultimately, our findings suggest the value of specific HMO blends to alleviate pain associated FGIDs such as infantile colic or Irritable Bowel Syndrome.


Assuntos
Dor Abdominal/dietoterapia , Disbiose/dietoterapia , Microbioma Gastrointestinal , Leite Humano/metabolismo , Oligossacarídeos/metabolismo , Dor Abdominal/metabolismo , Dor Abdominal/microbiologia , Dor Abdominal/psicologia , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Bactérias/metabolismo , Disbiose/metabolismo , Disbiose/microbiologia , Disbiose/psicologia , Fezes/microbiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oligossacarídeos/análise , Estresse Psicológico
9.
Am J Physiol Gastrointest Liver Physiol ; 300(6): G976-87, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21350188

RESUMO

Wound healing of the gastrointestinal mucosa is essential for the maintenance of gut homeostasis and integrity. Enteric glial cells play a major role in regulating intestinal barrier function, but their role in mucosal barrier repair remains unknown. The impact of conditional ablation of enteric glia on dextran sodium sulfate (DSS)-induced mucosal damage and on healing of diclofenac-induced mucosal ulcerations was evaluated in vivo in GFAP-HSVtk transgenic mice. A mechanically induced model of intestinal wound healing was developed to study glial-induced epithelial restitution. Glial-epithelial signaling mechanisms were analyzed by using pharmacological inhibitors, neutralizing antibodies, and genetically engineered intestinal epithelial cells. Enteric glial cells were shown to be abundant in the gut mucosa, where they associate closely with intestinal epithelial cells as a distinct cell population from myofibroblasts. Conditional ablation of enteric glia worsened mucosal damage after DSS treatment and significantly delayed mucosal wound healing following diclofenac-induced small intestinal enteropathy in transgenic mice. Enteric glial cells enhanced epithelial restitution and cell spreading in vitro. These enhanced repair processes were reproduced by use of glial-conditioned media, and soluble proEGF was identified as a secreted glial mediator leading to consecutive activation of epidermal growth factor receptor and focal adhesion kinase signaling pathways in intestinal epithelial cells. Our study shows that enteric glia represent a functionally important cellular component of the intestinal epithelial barrier microenvironment and that the disruption of this cellular network attenuates the mucosal healing process.


Assuntos
Enterite/enzimologia , Fator de Crescimento Epidérmico/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Mucosa Intestinal/enzimologia , Intestino Delgado/enzimologia , Neuroglia/enzimologia , Úlcera Péptica/enzimologia , Precursores de Proteínas/metabolismo , Cicatrização , Análise de Variância , Animais , Células CACO-2 , Forma Celular , Técnicas de Cocultura , Meios de Cultivo Condicionados/metabolismo , Sulfato de Dextrana , Diclofenaco , Modelos Animais de Doenças , Enterite/induzido quimicamente , Enterite/genética , Enterite/patologia , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Receptores ErbB/metabolismo , Quinase 1 de Adesão Focal/genética , Proteína Glial Fibrilar Ácida , Humanos , Mucosa Intestinal/patologia , Intestino Delgado/patologia , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/patologia , Comunicação Parácrina , Úlcera Péptica/induzido quimicamente , Úlcera Péptica/genética , Úlcera Péptica/patologia , Fosforilação , Interferência de RNA , Ratos , Transdução de Sinais , Simplexvirus/enzimologia , Simplexvirus/genética , Timidina Quinase/genética , Timidina Quinase/metabolismo , Fatores de Tempo , Transfecção
10.
Gastroenterology ; 138(5): 1772-82, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20152836

RESUMO

BACKGROUND & AIMS: Little is known about the environmental and nutritional regulation of the enteric nervous system (ENS), which controls gastrointestinal motility. Short-chain fatty acids (SCFAs) such as butyrate regulate colonic mucosa homeostasis and can modulate neuronal excitability. We investigated their effects on the ENS and colonic motility. METHODS: Effects of butyrate on the ENS were studied in colons of rats given a resistant starch diet (RSD) or intracecal perfusion of SCFAs. Effects of butyrate were also studied in primary cultures of ENS. The neurochemical phenotype of the ENS was analyzed with antibodies against Hu, choline acetyltransferase (ChAT), and neuronal nitric oxide synthase (nNOS) and by quantitative polymerase chain reaction. Signaling pathways involved were analyzed by pharmacologic and molecular biology methods. Colonic motility was assessed in vivo and ex vivo. RESULTS: In vivo and in vitro, RSD and butyrate significantly increased the proportion of ChAT- but not nNOS-immunoreactive myenteric neurons. Acetate and propionate did not reproduce the effects of butyrate. Enteric neurons expressed monocarboxylate transporter 2 (MCT2). Small interfering RNAs silenced MCT2 and prevented the increase in the proportion of ChAT- immunoreactive neurons induced by butyrate. Butyrate and trichostatin A increased histone H3 acetylation in enteric neurons. Effects of butyrate were prevented by inhibitors of the Src signaling pathway. RSD increased colonic transit, and butyrate increased the cholinergic-mediated colonic circular muscle contractile response ex vivo. CONCLUSION: Butyrate or histone deacetylase inhibitors might be used, along with nutritional approaches, to treat various gastrointestinal motility disorders associated with inhibition of colonic transit.


Assuntos
Butiratos/administração & dosagem , Colo/inervação , Sistema Nervoso Entérico/efeitos dos fármacos , Motilidade Gastrointestinal/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Acetilação , Animais , Células Cultivadas , Colina O-Acetiltransferase/genética , Colina O-Acetiltransferase/metabolismo , Colo/microbiologia , Carboidratos da Dieta/metabolismo , Relação Dose-Resposta a Droga , Sistema Nervoso Entérico/citologia , Sistema Nervoso Entérico/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Histonas/metabolismo , Hidroxiureia/metabolismo , Masculino , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Neurônios/metabolismo , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo I , Fenótipo , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
11.
FASEB J ; 24(4): 1082-94, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19906678

RESUMO

Enteric glial cells (EGCs) are essential in the control of gastrointestinal functions. Although lesions of EGCs are associated with neuronal degeneration in animal models, their direct neuroprotective role remains unknown. Therefore, the aims of this study were to demonstrate the direct neuroprotective effects of EGCs and to identify putative glial mediators involved. First, viral targeted ablation of EGCs in primary cultures of enteric nervous system increased neuronal death both under basal conditions and in the presence of oxidative stress (dopamine, hydrogen peroxide). Second, direct or indirect coculture experiments of EGC lines with primary cultures of enteric nervous system or neuroblastoma cell lines (SH-SY5Y) prevented neurotoxic effects induced by oxidative stress (increased membrane permeability, release of neuronal specific enolase, caspase-3 immunoreactivity, changes in [Ca(2+)](i) response). Finally, combining pharmacological inhibition and mRNA silencing methods, we demonstrated that neuroprotective effects of EGCs were mediated in part by reduced glutathione but not by oxidized glutathione or by S-nitrosoglutathione. Our study identified the neuroprotective effects of EGCs via their release of reduced glutathione, extending their critical role in physiological contexts and in enteric neuropathies.-Abdo, H., Derkinderen, P., Gomes, P., Chevalier, J., Aubert, P., Masson, D., Galmiche, J.-P., Vanden Berghe, P., Neunlist, M., Lardeux, B. Enteric glial cells protect neurons from oxidative stress in part via reduced glutathione.


Assuntos
Trato Gastrointestinal/metabolismo , Glutationa/metabolismo , Neurônios/metabolismo , Estresse Oxidativo , Animais , Cálcio/metabolismo , Caspase 3/genética , Caspase 3/metabolismo , Linhagem Celular Tumoral , Dopamina/farmacologia , Feminino , Trato Gastrointestinal/inervação , Inativação Gênica/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuroglia , Oxidantes/farmacologia , Oxirredução/efeitos dos fármacos , Fosfopiruvato Hidratase/genética , Fosfopiruvato Hidratase/metabolismo , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
12.
J Neurochem ; 115(3): 694-706, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20731759

RESUMO

Accumulated evidence emphasizes the importance of α-synuclein expression levels in Parkinson's disease (PD) pathogenesis. PD is a multicentric disorder that affects the enteric nervous system (ENS), whose involvement may herald the degenerative process in the CNS. We therefore undertook the present study to investigate the mechanisms involved in the regulation of expression of α-synuclein in the ENS. The regulation of α-synuclein expression was assessed by qPCR and western blot analysis in rat primary culture of ENS treated with KCl and forskolin. A pharmacological approach was used to decipher the signaling pathways involved. Intraperitoneal injections of Bay K-8644 and forskolin were performed in mice, whose proximal colons were further analyzed for α-synuclein expression. Depolarization and forskolin increased α-synuclein mRNA and protein expression in primary cultures of ENS, although L-type calcium channel and protein kinase A, respectively. Both stimuli increased α-synuclein expression through a Ras/extracellular signal-regulated kinases pathway. An increase in α-synuclein expression was also observed in vivo in the ENS of mice injected with Bay K-8644 or forskolin. In conclusion, we have identified stimuli leading to α-synuclein over-expression in the ENS, which could be critical in the initiation of the pathological process in PD.


Assuntos
AMP Cíclico/farmacologia , Sistema Nervoso Entérico/metabolismo , Neurônios/metabolismo , alfa-Sinucleína/biossíntese , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Animais , Western Blotting , Agonistas dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Colforsina/farmacologia , Eletrofisiologia , Sistema Nervoso Entérico/citologia , Sistema Nervoso Entérico/efeitos dos fármacos , Feminino , Imuno-Histoquímica , Indicadores e Reagentes , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Fosfopiruvato Hidratase/metabolismo , Gravidez , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas ras/metabolismo
13.
Obes Surg ; 27(10): 2590-2598, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28429171

RESUMO

BACKGROUND: Increased lipopolysaccharide (LPS) translocation due to altered intestinal permeability has been suggested as a mechanism for obesity-associated insulin resistance. The goal of this study was to assess the effect of sleeve gastrectomy (SG) on intestinal barrier permeability in diet-induced obese mice. MATERIALS AND METHODS: Four weeks after surgery, the effects of SG on intestinal permeabilities were assessed ex vivo and in vivo in male C57Bl/6J mice fed a high-fat diet. Gene expression of tight junction proteins and inflammatory cytokines was measured in jejunum, colon, liver, and inguinal adipose tissue. Plasma LPS was quantified by HPLCMS/MS spectrometry. RESULTS: SG significantly reduced body weight and improved glucose homeostasis, as expected. SG decreased paracellular (p = 0.01) and transcellular permeability (p = 0.03) in the jejunum; and increased mRNA levels of the tight junction proteins Jam A (p = 0.02) and occludin (p = 0.01). In contrast in the distal colon, paracellular permeability tended to be increased (p = 0.07) while transcellular permeability was significantly induced (p = 0.03) after SG. In vivo, the paracellular permeability was significantly increased 3 weeks after SG (p = 0.02). Plasma LPS level were increased after SG (p = 0.03), as well as mRNA levels of adipose and hepatic inflammatory markers (p = 0.02). CONCLUSIONS: SG significantly modifies intestinal permeability in a differential manner between the proximal and distal intestine. These changes promote LPS translocation in plasma, induce a low-grade pro-inflammatory state in adipose tissue and liver, but do not impair the SG-induced glucose homeostasis improvement.


Assuntos
Dieta Hiperlipídica , Mucosa Intestinal/metabolismo , Obesidade/cirurgia , Tecido Adiposo/metabolismo , Adiposidade , Animais , Gastrectomia/métodos , Resistência à Insulina , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/etiologia , Permeabilidade
14.
Hum Mutat ; 25(5): 468-75, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15832307

RESUMO

Single nucleotide primer extension reaction has been widely used in DNA testing, and several detection methods based on this core allelic discrimination have been developed. Most of the reported formats are based on a two step protocol involving first, a liquid phase extension reaction, then a physical separation process (chromatography, electrophoresis, capture on solid support, mass spectrometry). Here we describe a new strategy based on homogeneous time-resolved fluorescence (HTRF), which does not involve any separation process and which allows a simple "mix and measure" protocol. In this approach, a 5'-(europium) cryptate-labeled primer is elongated by a biotinylated dideoxynucleoside-triphosphate, followed by the addition of a streptavidin-acceptor conjugate, which gives rise to a long-life fluorescence resonance energy transfer (FRET) signal between the cryptate donor and the acceptor. We present the development of HTRF technology as applied to the diagnosis of tumor suppressor gene p53 (TP53) mutations, and its application to the analysis of genomic DNA from human tumoral samples. The sensitivity of the reported method is compared to the corresponding fluorescent polarization assay.


Assuntos
Análise Mutacional de DNA/métodos , Transferência Ressonante de Energia de Fluorescência/métodos , Linhagem Celular Tumoral , Polarização de Fluorescência/métodos , Genes p53/genética , Humanos , Mutação , Polimorfismo de Nucleotídeo Único
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA