Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Pharm ; 661: 124353, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38909926

RESUMEN

Labrafac™ MC60 (glycerol monocaprylocaprate) is a lipid-based excipient used in oral formulations as a solubiliser. Due to the high proportions of established permeability enhancers, caprylate (C8) and caprate (C10), in Labrafac™ MC60, we hypothesised that it might behave as an intestinal permeation enhancer. We therefore evaluated this using two paracellular markers (ex vivo) and insulin (in vivo) as model molecules. Ex vivo studies were conducted in isolated muscle-stripped rat colonic mucosae mounted in Ussing chambers. Apical addition of Labrafac™ MC60 (8, 12, and 16 mg/ml) enhanced the apparent permeability coefficients (Papp) of [14C] mannitol and FITC-dextran 4 kDa (FD4) across colonic mucosae. Similar effects were observed in isolated jejunal mucosae, but at higher concentrations (40 mg/ml). The enhancing capacity of Labrafac™ MC60 was transient due to reversibility of reductions in transepithelial electrical resistance (TEER) upon wash-out and effects on fluxes were molecular weight-dependent (MW) as suggested by fluxes of a set of high MW FITC-dextrans. The permeability enhancing effects of Labrafac™ MC60 ex vivo were maintained in the presence of simulated intestinal fluids, FaSSIF and FaSSCoF, in both jejunal and colonic mucosae, respectively. Following intra-intestinal regional instillations to rats, the relative bioavailability of 50 IU/kg insulin ad-mixed with Labrafac™ MC60 was 5 % in jejunum (40 mg/ml) and 6 % in colon (8 mg/ml). When Labrafac™ MC60 was combined with PEG-60 hydrogenated castor oil (1 % v/v), this further increased the bioavailability of insulin to 8 % in jejunum. Absorption enhancement was also maintained in the presence of FaSSIF in jejunal instillations. Histology after 120 min exposure to Labrafac™ MC60 in vivo for both jejunum and colon was similar to untreated control. Labrafac™ MC60 therefore acts as a non-damaging intestinal permeation enhancer for macromolecules and can be considered as another excipient in screening programmes to develop orally administered macromolecules.


Asunto(s)
Dextranos , Excipientes , Fluoresceína-5-Isotiocianato , Glicéridos , Absorción Intestinal , Mucosa Intestinal , Permeabilidad , Animales , Masculino , Absorción Intestinal/efectos de los fármacos , Dextranos/farmacocinética , Dextranos/administración & dosificación , Excipientes/química , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de los fármacos , Glicéridos/química , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/farmacocinética , Fluoresceína-5-Isotiocianato/administración & dosificación , Insulina , Ratas , Manitol , Ratas Wistar , Colon/metabolismo , Colon/efectos de los fármacos , Yeyuno/metabolismo , Yeyuno/efectos de los fármacos
2.
Tissue Cell ; 87: 102343, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38442546

RESUMEN

The hyperpermeability of intestinal epithelium is a key contributor to the occurrence and development of systemic inflammation. Although D-beta-hydroxybutyrate (BHB) exhibits various protective effects, whether it affects the permeability of intestinal epithelium in systemic inflammation has not been clarified. In this study, we investigated the effects of BHB on the intestinal epithelial permeability, the epithelial marker E-cadherin and the tight junction protein Claudin-1 in colon in the lipopolysaccharide (LPS)-induced systemic inflammation mouse model. Intraperitoneal injection of LPS was used to induce systemic inflammation and BHB was given by oral administration. The permeability of intestinal epithelium, the morphological changes of colonic epithelium, the distribution and generation of colon E-cadherin, and the Claudin-1 generation and its epithelial distribution in colon were detected. The results confirmed the intestinal epithelial hyperpermeability and inflammatory changes in colonic epithelium, with disturbed E-cadherin distribution in LPS-treated mice. Besides, colon Claudin-1 generation was decreased and its epithelial distribution in colon was weakened in LPS-treated mice. However, BHB treatments alleviated the LPS-induced hyperpermeability of intestinal epithelium, attenuated the colonic epithelial morphological changes and promoted orderly distribution of E-cadherin in colon. Furthermore, BHB up-regulated colon Claudin-1 generation and promoted its colonic epithelial distribution and content in LPS-treated mice. In conclusion, BHB may alleviate the hyperpermeability of intestinal epithelium via up-regulation of Claudin-1 in colon in LPS-treated mice.


Asunto(s)
Inflamación , Lipopolisacáridos , Ratones , Animales , Claudina-1 , Lipopolisacáridos/toxicidad , Ácido 3-Hidroxibutírico/farmacología , Cadherinas/metabolismo
3.
Prostate ; 84(6): 513-524, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38353479

RESUMEN

BACKGROUND: Gut microbiome is a community of microorganisms that lives in the human intestine and exerts various functions on the host, including metabolic, immunoregulatory, and control over cell proliferation. Gut microbiome alterations have been associated with various pathological conditions, such as diabetes mellitus, obesity, and cardiovascular diseases. Gut-prostate axis is explained by the association between gut microbiome quantitative and functional alterations along with increased intestinal epithelial permeability with prostatediseases. However, the pathophysiological mechanisms and clinical importance of this association are not completely clarified yet. METHODS: We conducted a narrative review of the most relevant articles in the Medline (US National Library of Medicine, Bethesda, MD, USA), Scopus (Elsevier, Amsterdam, The Netherlands) and Web of Science Core Collection (Thomson Reuters, Toronto, ON, Canada) databases. No chronological restrictions were applied, and the most related papers published until December 2023 were included. RESULTS: Gut microbiota (GM) and its metabolites are capable of modifying host androgen level, as well as prostate cancer (PCa) therapy response. Moreover, patients with inflammatory bowel disease have higher rates of prostatitis-like symptoms and a potential risk of developing PCa. CONCLUSIONS: There is evidence that interventions on the GM and its metabolites have a high potential to serve as diagnostic and therapeutic tools for prostate diseases, including PCa.


Asunto(s)
Diabetes Mellitus , Microbioma Gastrointestinal , Neoplasias de la Próstata , Prostatitis , Masculino , Humanos , Próstata/metabolismo , Microbioma Gastrointestinal/fisiología
4.
J Nutr Sci Vitaminol (Tokyo) ; 69(3): 164-175, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37394421

RESUMEN

The gastrointestinal (GI) tract is a series of hollow organs that play roles in food digestion and nutrient absorption. To perform these functions, they should recognize the luminal environment and elicit adequate physiological responses, including digestive juice secretion, peristaltic movements, etc. The Ussing chamber technique is an electrophysiological method for measuring transepithelial ion transport and permeability as short-circuit current (Isc) and transepithelial electrical tissue conductance (Gt) or resistance (TEER), respectively, in vitro. This technique can be applied for the measurement of luminal nutrient sensing and absorption. This article introduces practical methods for measuring luminal nutrient sensing and absorption using intestinal mucosa specimens isolated from humans and experimental animals.


Asunto(s)
Alimentos , Mucosa Intestinal , Animales , Humanos , Transporte Biológico , Mucosa Intestinal/metabolismo , Permeabilidad , Nutrientes
5.
Nanomaterials (Basel) ; 13(3)2023 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-36770503

RESUMEN

The development of oral drug delivery systems is challenging, and issues related to the mucus layer and low intestinal epithelial permeability have not yet been surmounted. The purpose of this study was to develop a promising formulation that is more adapted to in vivo absorption and to facilitate the administration of oral liraglutide. Cationic liposomes (CLs) linked to AT-1002 were prepared using a double-emulsion method, and BSA was adsorbed on the surface of the AT-CLs, resulting in protein corona cationic liposomes with AT-1002 (Pc-AT-CLs). The preparation method was determined by investigating various process parameters. The particle size, potential, and encapsulation efficiency (EE%) of the Pc-AT-CLs were 202.9 ± 12.4 nm, 1.76 ± 4.87 mV, and 84.63 ± 5.05%, respectively. The transmission electron microscopy (TEM) imaging revealed a nearly spherical structure of the Pc-AT-CLs, with a recognizable coating. The circular dichroism experiments confirmed that the complex preparation process did not affect the secondary structure of liraglutide. With the addition of BSA and AT-1002, the mucosal accumulation of the Pc-AT-CLs was nearly two times lower than that of the AT-CLs, and the degree of enteric metaplasia was 1.35 times higher than that of the PcCLs. The duration of the intestinal absorption of the Pc-AT-CLs was longer, offering remarkable biological safety.

6.
J Sci Food Agric ; 103(3): 1550-1560, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36208473

RESUMEN

BACKGROUND: Lipopolysaccharides (LPS) are the main pathogenic substances in Gram-negative bacteria. The aim of this study was to investigate the preventive effects of dietary curcumin (CUR) on LPS toxicity in the duck ileum. The duck diet was supplemented with CUR (0.5 g kg-1 ) for 28 days, while the birds were injected with LPS (0.5 mg kg-1 body weight per injection, administered as seven injections in the last week of the experimental period). RESULTS: LPS significantly decreased the ileal villus-to-crypt ratio in the non-supplemented CUR group. Dietary CUR alleviated LPS-induced morphological damage to the ileum. Moreover, dietary CUR alleviated oxidative stress by increasing the levels of total superoxide dismutase (T-SOD) (P < 0.05) and glutathione S-transferase (GST) (P < 0.05) and decreasing the production of malonic dialdehyde (MDA) (P < 0.05) in control ducks and LPS-challenged ducks. Dietary CUR significantly inhibited the LPS-induced massive production of inflammatory factors (IL-1ß, IL-6, and TNF-α) (P < 0.05). CUR induced the inhibition of TLR4 and activation of Nrf2 to reduce the expression of inflammation-related genes (TLR4, NF-κB, IKK, TXNIP, NLRP3, caspase-1, IL-1ß, IL-6, and TNF-α). Moreover, dietary CUR ameliorated the decrease in claudin-1 and occludin expression (P < 0.05) and improved ZO-1 expression in the duck ileum (P < 0.05). CONCLUSION: In conclusion, dietary CUR has beneficial effects on LPS-induced ileal damage, oxidative damage, and inflammatory response by inhibiting the TLR/NF-κB and activating the Nrf2 signaling pathways in ducks. This study provides valuable information regarding the therapeutic uses of CUR in duck ileitis. © 2022 Society of Chemical Industry.


Asunto(s)
Curcumina , Ileítis , Animales , Lipopolisacáridos/efectos adversos , Patos/metabolismo , Curcumina/farmacología , Curcumina/uso terapéutico , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/uso terapéutico , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/prevención & control , Estrés Oxidativo , Ileítis/inducido químicamente , Ileítis/genética , Ileítis/prevención & control
7.
Chronobiol Int ; 39(6): 872-885, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35232303

RESUMEN

Circadian rhythm disturbances induced by rotating shift work contribute to development of metabolic disorders. However, their effects on intestinal parameters such as epithelial permeability and fecal short chain fatty acid (SCFA) levels have not been established yet. This study was planned to investigate the changes in intestinal integrity, fecal SCFA levels, gut microbiota and nutritional intake of rotational shift workers. The study was conducted on ten male rotational shift workers, 25-40 years old. Circadian rhythm disruption was assumed to have occurred after 14 days in the night shift. Dietary data which was obtained by using 24 h record for 7 days, physical activity data, anthropometric measurements, fecal and blood samples were collected during day and night shift. Changes in dietary consumption, anthropometric measurements, blood chemistry and intestinal epithelial permeability indicator according to day and night shifts were not significant (p > .05). Additionally, acetic, propionic and total SCFA were associated with the intestinal permeability biomarker in night shift, but not in day shift (p < .05). Consumption of dark green vegetables and beans and peas was positively associated with fecal isobutyric acid and fecal total SCFA concentration (r = 0.685, p = .029; r = 0.695, p = .026, respectively). The proportions of the genus including Blautia, Bifidobacterium, Dialister, and Ruminococcus gnavus group increased when individuals shifted to the night shift. Gut microbiota changes responding to circadian rhythm disruption became more prominent when consumed high sugar diet. So, changes have been observed in the gut microbiota of rotational shift workers, especially in individuals with certain dietary pattern. Moreover, in individuals with the circadian rhythm disruption SCFAs levels have been demonstrated to be associated with intestinal barrier integrity. A better understanding of the relation among fecal SCFAs, gut microbiota, intestinal epithelial permeability and circadian rhythm disruption is necessary for the development of new dietary strategies for gut health.


Asunto(s)
Microbioma Gastrointestinal , Horario de Trabajo por Turnos , Adulto , Ritmo Circadiano , Dieta , Humanos , Masculino
8.
Adv Drug Deliv Rev ; 177: 113925, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34418495

RESUMEN

The use of chemical permeation enhancers (PEs) is the most widely tested approach to improve oral absorption of low permeability active agents, as represented by peptides. Several hundred PEs increase intestinal permeability in preclinical bioassays, yet few have progressed to clinical testing and, of those, only incremental increases in oral bioavailability (BA) have been observed. Still, average BA values of ~1% were sufficient for two recent FDA approvals of semaglutide and octreotide oral formulations. PEs are typically screened in static in vitro and ex-vivo models where co-presentation of active agent and PE in high concentrations allows the PE to alter barrier integrity with sufficient contact time to promote flux across the intestinal epithelium. The capacity to maintain high concentrations of co-presented agents at the epithelium is not reached by standard oral dosage forms in the upper GI tract in vivo due to dilution, interference from luminal components, fast intestinal transit, and possible absorption of the PE per se. The PE-based formulations that have been assessed in clinical trials in either immediate-release or enteric-coated solid dosage forms produce low and variable oral BA due to these uncontrollable physiological factors. For PEs to appreciably increase intestinal permeability from oral dosage forms in vivo, strategies must facilitate co-presentation of PE and active agent at the epithelium for a sustained period at the required concentrations. Focusing on peptides as examples of a macromolecule class, we review physiological impediments to optimal luminal presentation, discuss the efficacy of current PE-based oral dosage forms, and suggest strategies that might be used to improve them.


Asunto(s)
Composición de Medicamentos , Absorción Intestinal , Excipientes Farmacéuticos , Animales , Formas de Dosificación , Sistemas de Liberación de Medicamentos , Interacciones Alimento-Droga , Humanos , Permeabilidad , Excipientes Farmacéuticos/administración & dosificación , Excipientes Farmacéuticos/química , Excipientes Farmacéuticos/farmacocinética
9.
Tissue Barriers ; 9(2): 1890526, 2021 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-33660567

RESUMEN

The non-receptor protein tyrosine kinase 2ß (Pyk2) phosphorylated tricellular tight junction (tTJ) molecules angulin-1/LSR and tricellulin (TRIC) and the inhibitor PF-431396 (PF43) suppress angulin-1/LSR and TRIC recruitment to tTJs. The disruption of the intestinal epithelial barrier by high mobility group box 1 (HMGB1) and the inflammatory cytokines TNFα and IFNγ contributes to downregulation of angulin-1/LSR and TRIC in 2.5D culture of Caco-2 cells as a novel model of inflammatory bowel disease (IBD). In the present study, to investigate the roles of Pyk2 phosphorylated angulin-1/LSR and TRIC in the intestinal epithelial barrier, 2D and 2.5D cultures of Caco-2 cells were treated with the Pyk2 inhibitor PF-43 with or without HMGB1, inflammatory cytokines TNFα and IFNγ. Treatment with PF-43 increased expression of angulin-1/LSR, phosphorylated AMPK and phosphorylated MAPK and decreased that of phosphorylated JNK, with upregulation of the epithelial barrier and cellular metabolism measured as basal oxygen consumption rate (OCR) and ATP production in 2D culture. Treatment with PF-43 prevented the downregulation of the epithelial barrier by HMGB1 and inflammatory cytokines in 2D culture. Treatment with PF-43 prevented the epithelial hyperpermeability induced by HMGB1 and inflammatory cytokines in 2.5D culture. In 2.5D culture, treatment with PF-43 inhibited the decreases of angulin-1/LSR, TRIC, pJNK, pAMPK and pMAPK induced by HMGB1 and the inflammatory cytokines. Treatment with PF-43 inhibited in part the induced phosphorylation of the serine of angulin-1/LSR and TRIC. Pyk2 inhibitor PF-43 may have potential for use in therapy for IBD via its actions with regard to phosphorylated tTJs and cellular metabolism.


Asunto(s)
Citocinas/metabolismo , Células Epiteliales/metabolismo , Quinasa 2 de Adhesión Focal/antagonistas & inhibidores , Proteína HMGB1/metabolismo , Inflamación/metabolismo , Células CACO-2 , Línea Celular Tumoral , Humanos
10.
Int J Mol Sci ; 21(22)2020 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-33182652

RESUMEN

High mobility group box 1 protein (HMGB1) is involved in the pathogenesis of inflammatory bowel disease (IBD). Patients with IBD develop zinc deficiency. However, the detailed roles of HMGB1 and zinc deficiency in the intestinal epithelial barrier and cellular metabolism of IBD remain unknown. In the present study, Caco-2 cells in 2D culture and 2.5D Matrigel culture were pretreated with transforming growth factor-ß (TGF-ß) type 1 receptor kinase inhibitor EW-7197, epidermal growth factor receptor (EGFR) kinase inhibitor AG-1478 and a TNFα antibody before treatment with HMGB1 and inflammatory cytokines (TNFα and IFNγ). EW-7197, AG-1478 and the TNFα antibody prevented hyperpermeability induced by HMGB1 and inflammatory cytokines in 2.5D culture. HMGB1 affected cilia formation in 2.5D culture. EW-7197, AG-1478 and the TNFα antibody prevented the increase in cell metabolism induced by HMGB1 and inflammatory cytokines in 2D culture. Furthermore, ZnSO4 prevented the hyperpermeability induced by zinc chelator TPEN in 2.5D culture. ZnSO4 and TPEN induced cellular metabolism in 2D culture. The disruption of the epithelial barrier induced by HMGB1 and inflammatory cytokines contributed to TGF-ß/EGF signaling in Caco-2 cells. The TNFα antibody and ZnSO4 as well as EW-7197 and AG-1478 may have potential for use in therapy for IBD.


Asunto(s)
Citocinas/metabolismo , Etilenodiaminas/farmacología , Proteína HMGB1/metabolismo , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo , Compuestos de Anilina/farmacología , Células CACO-2 , Quelantes/farmacología , Proteína HMGB1/farmacología , Humanos , Mediadores de Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Interferón gamma/metabolismo , Interferón gamma/farmacología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Modelos Biológicos , Permeabilidad/efectos de los fármacos , Quinazolinas/farmacología , Receptores de Lipoproteína/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Triazoles/farmacología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/farmacología , Tirfostinos/farmacología , Sulfato de Zinc/farmacología
11.
J Microbiol Biotechnol ; 30(10): 1480-1487, 2020 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-32807750

RESUMEN

Our previous report determined that miR-144 is a key regulator of intestinal epithelial permeability in irritable bowel syndrome with diarrhea (IBS-D) rats. Recent evidence has shown that lactobacilli play an important role in the relief of IBS-D symptoms. However, few studies have addressed the mechanisms by which microRNAs and lactobacilli exert their beneficial effects on intestinal epithelial permeability. Hence, to elucidate whether miRNAs and lactobacilli play roles in intestinal epithelial barrier regulation, we compared miRNA expression levels in intestinal epithelial cells (IECs) under Lactobacillus casei (L. casei LC01) treatment. IECs and L. casei LC01 were co-cultured and then subjected to microRNA microarray assay. qRT-PCR, western blot and ELISA were used to detect the expression of occludin (OCLN) and zonula occludens 1 (ZO1/TJP1). The interaction between miRNAs and L. casei LC01 acting in IECs was investigated through transfection of RNA oligoribonucleotides and pcDNA 3.1 plasmid. The results are as follows: 1) L. casei LC01 decreased the expression of miR-144 and FD4 and promoted OCLN and ZO1 expression in IECs; 2) L. casei LC01 enhanced the barrier function of IECs via downregulation of miR-144 and upregulation of OCLN and ZO1; 3) Under L. casei LC01 treatment, OCLN and ZO1 overexpression could partially eliminate the promoting effect of miR-144 on intestinal permeability in IECs. Our results demonstrate that L. casei LC01 regulates intestinal permeability of IECs through miR-144 targeting of OCLN and ZO1. L. casei LC01 can be a possible therapeutic target for managing dysfunction of the intestinal epithelial barrier.


Asunto(s)
Lacticaseibacillus casei/metabolismo , MicroARNs/genética , Ocludina/genética , Proteína de la Zonula Occludens-1/genética , Animales , Línea Celular , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Humanos , Mucosa Intestinal/metabolismo , Intestinos , Síndrome del Colon Irritable/terapia , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Ocludina/metabolismo , Permeabilidad , Ratas , Proteína de la Zonula Occludens-1/metabolismo
12.
Nutrients ; 12(4)2020 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-32260563

RESUMEN

Obesity is characterized by fat accumulation, chronic inflammation and impaired satiety signaling, which may be due in part to gut microbial dysbiosis. Manipulations of the gut microbiota and its metabolites are attractive targets for obesity treatment. The predominant oligosaccharide found in human milk, acts as a prebiotic with beneficial effects on the host. However, little is known about the beneficial effects of 2'-FL in obesity. The aim of this study was to determine the beneficial effects of 2'-FL supplementation on the microbiota-gut-brain axis and the diet-induced obese phenotype in high fat (HF)-fed mice. Male C57/BL6 mice (n = 6/group; six weeks old) were counter-balanced into six weight-matched groups and fed either a low-fat (LF; 10% kcal as fat), HF (45% kcal as fat) or HF diet with 2'-FL (HF_2'-FL) at 1, 2, 5 and 10% (w/v) in drinking water for six weeks. General phenotypes (body weight, energy intake, fat and lean mass), cecal microbiome and metabolites, gut-brain signaling, intestinal permeability and inflammatory and lipid profiles were assessed. Only 10% 2'-FL, but not 1, 2 or 5%, decreased HF diet-induced increases in energy intake, fat mass and body weight gain. A supplementation of 10% 2'-FL changed the composition of cecal microbiota and metabolites compared to LF- and HF-fed mice with an increase in Parabacteroides abundance and lactate and pyruvate, respectively, whose metabolic effects corresponded to our study findings. In particular, 10% 2'-FL significantly reversed the HF diet-induced impairment of cholecystokinin-induced inhibition of food intake. Gene expressions of interleukin (IL)-1ß, IL-6, and macrophage chemoattractant protein-1 in the cecum were significantly downregulated by 10% 2'-FL compared to the HF group. Furthermore, 10% 2'-FL suppressed HF diet-induced upregulation of hepatic peroxisome proliferator-activated receptor gamma, a transcription factor for adipogenesis, at the gene level. In conclusion, 10% 2'-FL led to compositional changes in gut microbiota and metabolites associated with improvements in metabolic profiles and gut-brain signaling in HF-fed mice. These findings support the use of 2'-FL for modulating the hyperphagic response to HF diets and improving the microbiota-gut-brain axis.


Asunto(s)
Encéfalo/fisiología , Microbioma Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/fisiología , Obesidad/inducido químicamente , Trisacáridos/administración & dosificación , Alimentación Animal/análisis , Animales , Encéfalo/efectos de los fármacos , Dieta/veterinaria , Dieta Alta en Grasa , Suplementos Dietéticos , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Ratones , Obesidad/tratamiento farmacológico
13.
J Anim Physiol Anim Nutr (Berl) ; 103(4): 1174-1184, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30990939

RESUMEN

Eleutheroside B (EB) is a phenylpropanoid glycoside with anti-inflammatory properties, neuroprotective abilities, immunomodulatory effects, antinociceptive effects, and regulation of blood glucose. The aim of this study was to investigate the effects of EB on the barrier function in the intestinal porcine epithelial cells J2 (IPEC-J2). The IPEC-J2 cells were inoculated into 96-well plates at a density of 5 × 103 cells per well for 100% confluence. The cells were cultured in the presence of EB at concentrations of 0, 0.05, 0.10, and 0.20 mg/ml for 48 hr. Then, 0.10 mg/ml was selected as the suitable concentration for the estimation of transepithelial electric resistance (TEER) value, alkaline phosphatase activity, proinflammatory cytokines mRNA expression, tight junction mRNA and protein expression. The results of this study indicated that the supplementation of EB in IPEC-J2 cells decreased cellular membrane permeability and mRNA expression of proinflammatory cytokines, including interleukin-6 (IL-6), interferon-γ (INF-γ), and tumour necrosis factor-α (TNF-α). The supplementation of EB in IPEC-J2 cells increased tight junction protein expression and anti-inflammatory cytokines, interleukin 10 (IL-10) and transforming growth factor beta (TGF-ß). In addition, the western blotting and real-time quantitative polymerase chain reaction (RT-qPCR) results indicated that EB significantly (p < 0.05) increased the mRNA and protein expression of intestinal tight junction proteins, Claudin-3, Occludin, and Zonula Occludins protein-1 (ZO-1). Therefore, dietary supplementation of EB may increase intestinal barrier function, tight junction protein expression, anti-inflammatory cytokines, and decrease proinflammatory cytokines synthesis in IPEC-J2 cells.


Asunto(s)
Citocinas/metabolismo , Células Epiteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glucósidos/farmacología , Fenilpropionatos/farmacología , Porcinos , Proteínas de Uniones Estrechas/metabolismo , Animales , Línea Celular , Proliferación Celular , Citocinas/genética , Relación Dosis-Respuesta a Droga , Glucósidos/administración & dosificación , Yeyuno/citología , Fenilpropionatos/administración & dosificación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Uniones Estrechas/genética
14.
Cell Physiol Biochem ; 44(3): 1161-1173, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29179184

RESUMEN

BACKGROUND/AIMS: Intestinal permeability and stress have been implicated in the pathophysiology of irritable bowel syndrome (IBS). Cytokeratin 8 (CK8), for the first time, has been shown to mediate corticotropin-releasing factor (CRF)-induced changes in intestinal permeability in animal models of IBS. In this study, we investigated the regulatory effects of CRF on the permeability of human intestinal epithelial cells through the CK8-mediated tight junction. METHODS: The expression levels of corticotropin-releasing factor receptor 1 (CRFR1) and corticotropin-releasing factor receptor 2 (CRFR2) on the HT29 cell surface were determined by immunofluorescence, RT-PCR, and Western blotting. After treatment with 100 nM CRF for 72 h, the translocation of FITC-labelled dextran was measured in a transwell chamber; the structural changes of tight junctions were observed under transmission electron microscopy; the expression levels of CK8, F-actin and tight junction proteins ZO-1, claudin-1, and occludin were detected by immunoblotting and immunofluorescence. The activity of RhoA was detected by immunoprecipitation. Furthermore, the effects of CRF on intestinal epithelial permeability were examined in CK8-silenced HT29 cells, which were constructed by shRNA interference. RESULTS: CRF treatment increased FITC-labelled dextran permeability, caused the opening of tight junctions, induced increased fluorescence intensity of CK8 and decreased the intensities of ZO-1, claudin-1, and occludin, together with structural disruption. The expression levels of F-actin, occludin, claudin-1, and ZO-1 were downregulated. RhoA activity peaked at 30 min after CRF treatment. CRF-induced increased permeability, and downregulation of claudin-1 and occludin were not blocked by CK8 silencing. Nevertheless, CK8 silencing blocked the effects of CRF regarding the decrease in the expression levels of F-action and ZO-1 and increase in RhoA activity. CONCLUSION: CRF may increase intestinal epithelial permeability by upregulating CK8 expression, activating the RhoA signalling pathway, promoting intestinal epithelial actin remodelling, and decreasing the expression of the tight junction protein ZO-1. Other CK8-independent pathways may be involved in the downregulation of claudin-1 and occludin, which might also contribute to increased intestinal epithelial permeability.


Asunto(s)
Hormona Liberadora de Corticotropina/farmacología , Queratina-8/metabolismo , Permeabilidad/efectos de los fármacos , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Uniones Estrechas/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Actinas/metabolismo , Claudina-1/metabolismo , Células HT29 , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Queratina-8/antagonistas & inhibidores , Queratina-8/genética , Microscopía Electrónica , Microscopía Fluorescente , Ocludina/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Hormona Liberadora de Corticotropina/genética , Uniones Estrechas/metabolismo , Uniones Estrechas/ultraestructura , Proteína de la Zonula Occludens-1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
15.
Artículo en Inglés | MEDLINE | ID: mdl-28370715

RESUMEN

BACKGROUND: Intestinal epithelial barrier (IEB) dysfunction plays a critical role in various intestinal disorders affecting infants and children, including the development of food allergies and colitis. Recent studies highlighted the role of probiotics in regulating IEB functions and behavior in adults, but their effects in the newborn remain largely unknown. We therefore characterized in rat pups, the impact of Lactobacillus fermentum CECT 5716 (L. fermentum) on stress-induced IEB dysfunction, systemic immune response and exploratory behavior. METHODS: Newborn rats received daily by gavage either L. fermentum or water. Intestinal permeability to fluorescein sulfonic acid (FSA) and horseradish peroxidase (HRP) was measured following maternal separation (MS) and water avoidance stress (WAS). Immunohistochemical, transcriptomic, and Western blot analysis of zonula occludens-1 (ZO-1) distribution and expression were performed. Anxiety-like and exploratory behavior was assessed using the elevated plus maze test. Cytokine secretion of activated splenocytes was also evaluated. KEY RESULTS: L. fermentum prevented MS and WAS-induced IEB dysfunction in vivo. L. fermentum reduced permeability to both FSA and HRP in the small intestine but not in the colon. L. fermentum increased expression of ZO-1 and prevented WAS-induced ZO-1 disorganization in ileal epithelial cells. L. fermentum also significantly reduced stress-induced increase in plasma corticosteronemia. In activated splenocytes, L. fermentum enhanced IFNγ secretion while it prevented IL-4 secretion. Finally, L. fermentum increased exploratory behavior. CONCLUSIONS & INFERENCES: These results suggest that L. fermentum could provide a novel tool for the prevention and/or treatment of gastrointestinal disorders associated with altered IEB functions in the newborn.


Asunto(s)
Enfermedades Gastrointestinales/metabolismo , Mucosa Intestinal/metabolismo , Limosilactobacillus fermentum , Probióticos/administración & dosificación , Estrés Psicológico/complicaciones , Animales , Animales Recién Nacidos , Colon/metabolismo , Células Epiteliales/metabolismo , Conducta Exploratoria , Femenino , Enfermedades Gastrointestinales/complicaciones , Enfermedades Gastrointestinales/inmunología , Enfermedades Gastrointestinales/terapia , Privación Materna , Permeabilidad , Ratas Sprague-Dawley , Proteína de la Zonula Occludens-1/metabolismo
16.
Antioxidants (Basel) ; 5(4)2016 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-27845706

RESUMEN

Oxidative stress in the small intestinal epithelium can lead to barrier malfunction. In this study, the effect of rosmarinic acid (RA), quercetin (Que), gallic acid (GA), lipoic acid (LA), ethoxyquin (ETQ) and Se-methionine (SeMet) pre-treatments using 2 mM Trolox as a control on the viability and the generation of intracellular reactive oxygen species (iROS) of oxidatively (H2O2) stressed intestinal porcine epithelial cells (IPEC-J2) was investigated. A neutral red assay showed that RA (50-400 µM), Que (12.5-200 µM), GA (50-400 µM), ETQ (6.25-100 µM), and SeMet (125-1000 µM) pre-treatments but not LA significantly increased the viability of H2O2-stressed IPEC-J2 cells (p < 0.05). A 5-(and-6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate, acetyl ester (CM-H2DCFDA) fluorescent probe showed that RA (100-600 µM), Que (25-800 µM), ETQ (3.125-100 µM) and SeMet (500-2000 µM) pre-treatments significantly reduced iROS in IPEC-J2 monolayers (p < 0.05). Moreover, RA and Que were most effective in reducing iROS. Therefore, the effects of RA and Que on barrier functioning in vitro were examined. RA and Que pre-treatments significantly decreased fluorescein isothiocyanate (FITC)-conjugated dextran-4 (4 kDa) permeability and transepithelial electrical resistance (TEER) of an IPEC-J2 cell monolayer (p < 0.05). These in vitro results of RA and Que hold promise for their use as antioxidants in pig feed.

17.
Mol Cell Biochem ; 416(1-2): 23-32, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27023908

RESUMEN

The increased intestinal permeability and functional impairment play an important role in type 2 diabetes (T2D), and melatonin may possess enteroprotection properties. Therefore, we used streptozotocin-induced diabetic rat model to investigate the regulation of intestinal permeability by melatonin. Rats were randomly divided into three groups, including control, diabetes mellitus (DM), and DM rats treated with melatonin. Melatonin was administered (10 mg/kg/day) by gavage for 24 weeks. The DM rats significantly increased the serum fasting blood glucose and lipid levels, which were alleviated by melatonin treatment. Importantly, the intestinal epithelial permeability was significantly increased in DM rats but was ameliorated following treatment with melatonin. These findings also indicated the expression of myosin light chain kinase (MLCK) and phosphorylation of MLC targeting subunit (MYPT) induced myosin light chain (MLC) phosphorylation level was markedly elevated in hyperglycemic and hyperlipidemic status. They were partly associated with down-regulated membrane type 1 and 2 (MT1 and MT2) expression, and up-regulated Rho-associated protein kinase (ROCK) expression and increased extracellular signal-regulated kinase (ERK) phosphorylation. However, the changes in target protein expression were reversed by melatonin. In conclusion, our results show melatonin beneficial effects on impaired intestinal epithelial permeability in T2D by suppressing ERK/MLCK- and ROCK/MCLP-dependent MLC phosphorylation.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Absorción Intestinal/efectos de los fármacos , Melatonina/farmacocinética , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Animales , Diabetes Mellitus Experimental/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Masculino , Melatonina/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Melatonina MT1/metabolismo , Receptor de Melatonina MT2/metabolismo , Quinasas Asociadas a rho/metabolismo
18.
Physiol Rep ; 3(3)2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25780093

RESUMEN

Epithelial barrier dysfunction has been implicated as one of the major contributors to the pathogenesis of inflammatory bowel disease. The increase in intestinal permeability allows the translocation of luminal antigens across the intestinal epithelium, leading to the exacerbation of colitis. Thus, therapies targeted at specifically restoring tight junction barrier function are thought to have great potential as an alternative or supplement to immunology-based therapies. In this study, we screened Bifidobacterium, Enterococcus, and Lactobacillus species for beneficial microbes to strengthen the intestinal epithelial barrier, using the human intestinal epithelial cell line (Caco-2) in an in vitro assay. Some Bifidobacterium and Lactobacillus species prevented epithelial barrier disruption induced by TNF-α, as assessed by measuring the transepithelial electrical resistance (TER). Furthermore, live Bifidobacterium species promoted wound repair in Caco-2 cell monolayers treated with TNF-α for 48 h. Time course (1)H-NMR-based metabonomics of the culture supernatant revealed markedly enhanced production of acetate after 12 hours of coincubation of B. bifidum and Caco-2. An increase in TER was observed by the administration of acetate to TNF-α-treated Caco-2 monolayers. Interestingly, acetate-induced TER-enhancing effect in the coculture of B. bifidum and Caco-2 cells depends on the differentiation stage of the intestinal epithelial cells. These results suggest that Bifidobacterium species enhance intestinal epithelial barrier function via metabolites such as acetate.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...