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

Banco de datos
Tipo del documento
Publication year range
1.
Int J Mol Sci ; 24(8)2023 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-37108590

RESUMEN

The "leaky gut" syndrome describes a damaged (leaky) intestinal mucosa and is considered a serious contributor to numerous chronic diseases. Chronic inflammatory bowel diseases (IBD) are particularly associated with the "leaky gut" syndrome, but also allergies, autoimmune diseases or neurological disorders. We developed a complex in vitro inflammation-triggered triple-culture model using 21-day-differentiated human intestinal Caco-2 epithelial cells and HT29-MTX-E12 mucus-producing goblet cells (90:10 ratio) in close contact with differentiated human macrophage-like THP-1 cells or primary monocyte-derived macrophages from human peripheral blood. Upon an inflammatory stimulus, the characteristics of a "leaky gut" became evident: a significant loss of intestinal cell integrity in terms of decreased transepithelial/transendothelial electrical resistance (TEER), as well as a loss of tight junction proteins. The cell permeability for FITC-dextran 4 kDa was then increased, and key pro-inflammatory cytokines, including TNF-alpha and IL-6, were substantially released. Whereas in the M1 macrophage-like THP-1 co-culture model, we could not detect the release of IL-23, which plays a crucial regulatory role in IBD, this cytokine was clearly detected when using primary human M1 macrophages instead. In conclusion, we provide an advanced human in vitro model that could be useful for screening and evaluating therapeutic drugs for IBD treatment, including potential IL-23 inhibitors.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Macrófagos , Humanos , Células CACO-2 , Células THP-1 , Macrófagos/metabolismo , Inflamación/metabolismo , Citocinas/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo , Interleucina-23/metabolismo
2.
Br J Nutr ; 118(5): 321-332, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28901890

RESUMEN

We investigated the effects of dietary l-arginine level and feeding duration on the intestinal damage of broilers induced by Clostridium perfringens (CP) in vivo, and the antimicrobial effect of its metabolite nitric oxide (NO) in vitro. The in vivo experiment was designed as a factorial arrangement of three dietary treatments×two challenge statuses. Broilers were fed a basal diet (CON) or a high-arginine diet (ARG) containing 1·87 % l-arginine, or CON for the first 8 d and ARG from days 9 to 28 (CON/ARG). Birds were co-infected with or without Eimeria and CP (EM/CP). EM/CP challenge led to intestinal injury, as evidenced by lower plasma d-xylose concentration (P<0·01), higher paracellular permeability in the ileum (P<0·05) and higher numbers of Escherichia coli (P<0·05) and CP (P<0·001) in caecal digesta; however, this situation could be alleviated by l-arginine supplementation (P<0·05). The intestinal claudin-1 and occludin mRNA expression levels were decreased (P<0·05) following EM/CP challenge; this was reversed by l-arginine supplementation (P<0·05). Moreover, EM/CP challenge up-regulated (P<0·05) claudin-2, interferon-γ (IFN-γ), toll-like receptor 2 and nucleotide-binding oligomerisation domain 1 (NOD1) mRNA expression, and l-arginine supplementation elevated (P<0·05) IFN-γ, IL-10 and NOD1 mRNA expression. In vitro study showed that NO had bacteriostatic activity against CP (P<0·001). In conclusion, l-arginine supplementation could inhibit CP overgrowth and alleviate intestinal mucosal injury by modulating innate immune responses, enhancing barrier function and producing NO.


Asunto(s)
Arginina/administración & dosificación , Clostridium perfringens/efectos de los fármacos , Dieta/veterinaria , Inmunidad Innata , Mucosa Intestinal/efectos de los fármacos , Intestinos/microbiología , Alimentación Animal/análisis , Animales , Pollos , Claudina-1/genética , Claudina-1/metabolismo , Claudina-2/genética , Claudina-2/metabolismo , Suplementos Dietéticos , Eimeria/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Óxido Nítrico/metabolismo , Proteína Adaptadora de Señalización NOD1/genética , Proteína Adaptadora de Señalización NOD1/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Regulación hacia Arriba , Xilosa/sangre
3.
Br J Nutr ; 118(9): 661-672, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29185927

RESUMEN

Our previous study demonstrated that supplemental psyllium fibre increased cytoprotective heat-shock protein (Hsp) 25 levels in the intestinal cells of mice. Here, we examined the effect of psyllium fibre on colonic gene and protein expression and faecal microbiota in normal and colitic mice to improve the understanding of the preventive role of the supplement. DNA microarray analysis revealed that a 10 % psyllium fibre diet administered for 5 d up-regulated eleven extracellular matrix (ECM)-associated genes, including collagens and fibronectins, in normal mice. Acute colitis was induced using dextran sodium sulphate (DSS) in mice that were administered a pre-feeding 5 to 10 % psyllium fibre diet for 5 d. Psyllium fibre partially ameliorated or resolved the DSS-induced colon damage and inflammation characterised by body weight loss, colon shortening, increased levels of pro-inflammatory cytokines and decreased tight junction protein expression in the colon. Analysis of faecal microbiota using denaturing gradient gel electrophoresis of the PCR-amplified 16S rRNA gene demonstrated that psyllium fibre affected the colonic microbiota. Intestinal permeability was evaluated by growing intestinal Caco-2 cell monolayers on membrane filter supports coated with or without fibronectin and collagen. Cells grown on collagen and fibronectin coating showed higher transepithelial electrical resistance, indicating a strengthening of barrier integrity. Therefore, increased Hsp25 levels and modification of colonic ECM contribute to the observed psyllium-mediated protection against DSS-induced colitis. Furthermore, ECM modification appears to play a role in the strengthening of the colon barrier. In conclusion, psyllium fibre may be useful in the prevention of intestinal inflammatory diseases.


Asunto(s)
Colitis/tratamiento farmacológico , Fibras de la Dieta/farmacología , Inflamación/tratamiento farmacológico , Intestinos/efectos de los fármacos , Psyllium/farmacología , Animales , Células CACO-2 , Colitis/inducido químicamente , Colágeno/farmacología , Citocinas/sangre , ADN Bacteriano/aislamiento & purificación , Sulfato de Dextran , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Heces/microbiología , Fibronectinas/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Permeabilidad/efectos de los fármacos , ARN Ribosómico 16S/aislamiento & purificación , Uniones Estrechas/genética , Uniones Estrechas/metabolismo
4.
J Nutr ; 146(8): 1492-8, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27358412

RESUMEN

BACKGROUND: One promising strategy for reducing human salmonellosis induced by Salmonella Enteritidis is to supplement animal diets with natural feed additives such as mannan oligosaccharides (MOSs). OBJECTIVE: We sought to investigate the potential role of Salmosan (S-ßGM), an MOS product extremely rich in ß-galactomannan, in preventing epithelial barrier function disruption induced by S. Enteritidis colonization in an in vitro model of intestinal Caco-2 cells in culture. METHODS: Differentiated Caco-2 cells were incubated for 3 h with S. Enteritidis at a multiplicity of infection of 10 in the absence or presence of 500 µg S-ßGM/mL. Paracellular permeability (PP) was assessed by transepithelial electrical resistance (TER), d-mannitol, and fluorescein isothiocyanate-dextran (FD-4) flux. Tight junction proteins and cytoskeletal actin were also localized by confocal microscopy. Reactive oxygen species (ROS) and lipid peroxidation products were evaluated. Scanning and transmission electron microscopy were used to visualize S. Enteritidis adhesion to, and invasion of, the Caco-2 cell cultures. RESULTS: Compared with controls, TER was significantly reduced by 30%, and d-mannitol and FD-4 flux were significantly increased by 374% and 54% in S. Enteritidis-infected cultures, respectively. The presence of S-ßGM in infected cultures induced total recoveries of TER and FD-4 flux to values that did not differ from the control and a partial recovery of d-mannitol flux. These effects were confirmed by immunolocalization of actin, zonula occludens protein 1, and occludin. Similar results were obtained for Salmonella Dublin. The protection of S-ßGM on PP in infected cultures may be associated with a total recovery of ROS production to values that did not differ from the control. Moreover, S-ßGM has the capacity to agglutinate bacteria, leading to a significant reduction of 32% in intracellular S Enteritidis. CONCLUSION: The results demonstrate that S-ßGM contributes to protecting epithelial barrier function in a Caco-2 cell model disrupted by S. Enteritidis.


Asunto(s)
Colon/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mananos/farmacología , Oligosacáridos/farmacología , Infecciones por Salmonella/fisiopatología , Salmonella enteritidis , Uniones Estrechas/efectos de los fármacos , Actinas/metabolismo , Células CACO-2 , Colon/metabolismo , Colon/microbiología , Colon/fisiopatología , Suplementos Dietéticos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Galactosa/análogos & derivados , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/fisiopatología , Ocludina/metabolismo , Permeabilidad , Infecciones por Salmonella/microbiología , Proteína de la Zonula Occludens-1/metabolismo
5.
Br J Nutr ; 115(6): 984-93, 2016 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-26810899

RESUMEN

Whey protein concentrate (WPC) has been reported to have protective effects on the intestinal barrier. However, the molecular mechanisms involved are not fully elucidated. Transforming growth factor-ß1 (TGF-ß1) is an important component in the WPC, but whether TGF-ß1 plays a role in these processes is not clear. The aim of this study was to investigate the protective effects of WPC on the intestinal epithelial barrier as well as whether TGF-ß1 is involved in these protection processes in a piglet model after lipopolysaccharide (LPS) challenge. In total, eighteen weanling pigs were randomly allocated to one of the following three treatment groups: (1) non-challenged control and control diet; (2) LPS-challenged control and control diet; (3) LPS+5 %WPC diet. After 19 d of feeding with control or 5 %WPC diets, pigs were injected with LPS or saline. At 4 h after injection, pigs were killed to harvest jejunal samples. The results showed that WPC improved (P<0·05) intestinal morphology, as indicated by greater villus height and villus height:crypt depth ratio, and intestinal barrier function, which was reflected by increased transepithelial electrical resistance and decreased mucosal-to-serosal paracellular flux of dextran (4 kDa), compared with the LPS group. Moreover, WPC prevented the LPS-induced decrease (P<0·05) in claudin-1, occludin and zonula occludens-1 expressions in the jejunal mucosae. WPC also attenuated intestinal inflammation, indicated by decreased (P<0·05) mRNA expressions of TNF-α, IL-6, IL-8 and IL-1ß. Supplementation with WPC also increased (P<0·05) TGF-ß1 protein, phosphorylated-Smad2 expression and Smad4 and Smad7 mRNA expressions and decreased (P<0·05) the ratios of the phosphorylated to total c-jun N-terminal kinase (JNK) and p38 (phospho-JNK:JNK and p-p38:p38), whereas it increased (P<0·05) the ratio of extracellular signal-regulated kinase (ERK) (phospho-ERK:ERK). Collectively, these results suggest that dietary inclusion of WPC attenuates the LPS-induced intestinal injury by improving mucosal barrier function, alleviating intestinal inflammation and influencing TGF-ß1 canonical Smad and mitogen-activated protein kinase signalling pathways.


Asunto(s)
Suplementos Dietéticos , Modelos Animales de Enfermedad , Enterocolitis/prevención & control , Mucosa Intestinal/fisiopatología , Intestinos/fisiopatología , Proteínas de Uniones Estrechas/metabolismo , Proteína de Suero de Leche/uso terapéutico , Animales , Cruzamientos Genéticos , Citocinas/genética , Citocinas/metabolismo , Impedancia Eléctrica , Enterocolitis/metabolismo , Enterocolitis/patología , Enterocolitis/fisiopatología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Intestinos/inmunología , Intestinos/patología , Lipopolisacáridos/toxicidad , Sistema de Señalización de MAP Quinasas , Masculino , Orquiectomía/veterinaria , Permeabilidad , Distribución Aleatoria , Sus scrofa , Proteínas de Uniones Estrechas/genética , Factor de Crecimiento Transformador beta1/análisis , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/uso terapéutico , Destete , Proteína de Suero de Leche/química
6.
Gels ; 8(11)2022 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-36354594

RESUMEN

Therapeutic macromolecules (e.g., protein and peptide drugs) present bioavailability challenges via extravascular administration. The nasal route presents an alternative non-invasive route for these drugs, although low bioavailability remains challenging. Co-administration of permeation enhancers is a promising formulation approach to improve the delivery of poorly bioavailable drugs. The aim of this study was to prepare and characterize chitosan microparticulate formulations containing a macromolecular model compound (fluorescein isothiocyanate dextran 4400, FD-4) and a bioenhancer (piperine). Ionic gelation was used to produce chitosan microparticle delivery systems with two distinct microparticle sizes, differing one order of magnitude in size (±20 µm and ±200 µm). These two microparticle delivery systems were formulated into thermosensitive gels and their drug delivery performance was evaluated across ovine nasal epithelial tissues. Dissolution studies revealed a biphasic release pattern. Rheometry results demonstrated a sol-to-gel transition of the thermosensitive gel formulation at a temperature of 34 °C. The microparticles incorporating piperine showed a 1.2-fold increase in FD-4 delivery across the excised ovine nasal epithelial tissues as compared to microparticles without piperine. This study therefore contributed to advancements in ionic gelation methods for the formulation of particulate systems to enhance macromolecular nasal drug delivery.

7.
Methods Mol Biol ; 2321: 169-175, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34048016

RESUMEN

Gut barrier function has been hypothesized to play a critical role in the pathophysiology of sepsis. Measuring intestinal permeability allows for a determination of barrier dysfunction under conditions of health and disease. Fluorescence-conjugated dyes such as fluorescein isothiocyanate-4 kDa dextran (FD4) have been commonly used for evaluating hyperpermeability. Here we describe a common method to measure gut permeability in vivo, following gavage with different sized dyes. In addition, we describe an ex vivo everted gut sac model that allows for discrimination of permeability by segmental geographic location along the intestine.


Asunto(s)
Mucosa Intestinal/metabolismo , Sepsis/metabolismo , Animales , Dextranos/metabolismo , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Ratones , Permeabilidad
8.
Front Cell Dev Biol ; 9: 723927, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34409041

RESUMEN

Appendage development requires the coordinated function of signaling pathways and transcription factors to pattern the leg along the three main axes: the antero-posterior (AP), proximo-distal (PD), and dorso-ventral (DV). The Drosophila leg DV axis is organized by two morphogens, Decapentaplegic (Dpp), and Wingless (Wg), which direct dorsal and ventral cell fates, respectively. However, how these signals regulate the differential expression of its target genes is mostly unknown. In this work, we found that two members of the Drosophila forkhead family of transcription factors, Fd4 and Fd5 (also known as fd96Ca and fd96Cb), are identically expressed in the ventro-lateral domain of the leg imaginal disc in response to Dpp signaling. Here, we analyze the expression regulation and function of these genes during leg development. We have generated specific mutant alleles for each gene and a double fd4/fd5 mutant chromosome to study their function during development. We highlight the redundant role of the fd4/fd5 genes during the formation of the sex comb, a male specific structure that appears in the ventro-lateral domain of the prothoracic leg.

9.
Acta Pharm Sin B ; 11(9): 2859-2879, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34589401

RESUMEN

Parkinson's disease (PD) is the second most common neurodegenerative disease, but none of the current treatments for PD can halt the progress of the disease due to the limited understanding of the pathogenesis. In PD development, the communication between the brain and the gastrointestinal system influenced by gut microbiota is known as microbiota-gut-brain axis. However, the explicit mechanisms of microbiota dysbiosis in PD development have not been well elucidated yet. FLZ, a novel squamosamide derivative, has been proved to be effective in many PD models and is undergoing the phase I clinical trial to treat PD in China. Moreover, our previous pharmacokinetic study revealed that gut microbiota could regulate the absorption of FLZ in vivo. The aims of our study were to assess the protective effects of FLZ treatment on PD and to further explore the underlying microbiota-related mechanisms of PD by using FLZ as a tool. In the current study, chronic oral administration of rotenone was utilized to induce a mouse model to mimic the pathological process of PD. Here we revealed that FLZ treatment alleviated gastrointestinal dysfunctions, motor symptoms, and dopaminergic neuron death in rotenone-challenged mice. 16S rRNA sequencing found that PD-related microbiota alterations induced by rotenone were reversed by FLZ treatment. Remarkably, FLZ administration attenuated intestinal inflammation and gut barrier destruction, which subsequently inhibited systemic inflammation. Eventually, FLZ treatment restored blood-brain barrier structure and suppressed neuroinflammation by inhibiting the activation of astrocytes and microglia in the substantia nigra (SN). Further mechanistic research demonstrated that FLZ treatment suppressed the TLR4/MyD88/NF-κB pathway both in the SN and colon. Collectively, FLZ treatment ameliorates microbiota dysbiosis to protect the PD model via inhibiting TLR4 pathway, which contributes to one of the underlying mechanisms beneath its neuroprotective effects. Our research also supports the importance of microbiota-gut-brain axis in PD pathogenesis, suggesting its potential role as a novel therapeutic target for PD treatment.

10.
Int J Pharm ; 576: 118987, 2020 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-31870961

RESUMEN

A novel Liposome Aggregate Platform (LAP) system for prolonged retention of drugs in the posterior eye segment after intravitreal injection (IVT) was developed and evaluated. Calcein, FITC-dextran-4000 (FD4) and Flurbiprofen (FLB), were encapsulated in negatively charged liposomes, and mixed with protamine to produce the LAP. The lipid/protamine ratio was fixed, in order to have a convenient aggregation rate permitting IVT injection and also a sustained release of liposome-entrapped molecules (in vitro) from LAP. In vitro release studies confirmed the potential of LAP system consisted of HPC/DPPG/Chol liposomes and protamine (at 1:1 w/w to lipid), to delay calcein, FD4 and FLB release, compared to free liposomes. In vivo studies demonstrated increased vitreous retention of liposomes and LAP for all molecules, compared to the corresponding solutions; however the retention of FD4 is similar for non-aggregated liposomes and LAP, and calcein retention is only slightly increased by LAP compared to liposomes. The later result may be connected with the visible ocular inflammation caused by both dyes; interestingly inflammation was moderately reduced when dyes were entrapped in liposomes and even more when in LAP. No visible inflammation was demonstrated for FLB, and the LAP system significantly increased the retention of FLB in the ocular tissues (compared to non-aggregated liposomes). Taking into account the capability of the novel LAP system to decrease inflammatory reactions towards calcein and FD4, and prolong the retention of FLB in ocular tissues, it is concluded that such systems, after further optimization, may be considered as promising effective and safe approaches for treatment of posterior segment ocular pathologies.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Flurbiprofeno/administración & dosificación , Lípidos/química , Liposomas , Segmento Posterior del Ojo/metabolismo , Protaminas/química , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/farmacocinética , Preparaciones de Acción Retardada , Dextranos/administración & dosificación , Dextranos/química , Dextranos/metabolismo , Composición de Medicamentos , Liberación de Fármacos , Fluoresceína-5-Isotiocianato/administración & dosificación , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/química , Fluoresceína-5-Isotiocianato/metabolismo , Fluoresceínas/administración & dosificación , Fluoresceínas/química , Fluoresceínas/metabolismo , Flurbiprofeno/química , Flurbiprofeno/farmacocinética , Inyecciones Intravítreas , Modelos Biológicos , Conejos , Distribución Tisular
11.
Eur J Pharm Biopharm ; 130: 281-289, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30006244

RESUMEN

Nanoparticles (NP) only different in mucoadhesivity are compared for impact on drug oral bioavailability. Two polymeric NP types based on quaternary ammonium-chitosan (NP QA-Ch) and S-protected thiolated derivative thereof (NP QA-Ch-S-pro), respectively, containing the macromolecular drug model, FD4, were prepared by crosslinking each polymer with reduced MW hyaluronic acid. The structure of basic polymers was determined by H1NMR analysis. NP were similar in size (371 ±â€¯38 vs. 376 ±â€¯82 nm); polydispersity index (0.39 ±â€¯0.08 vs. 0.41 ±â€¯0.10); zeta potential (13.4 ±â€¯0.9 vs. 11.9 ±â€¯1.2 mV); reversible interactions with drug (bound drug, 67 vs. 66%); encapsulation efficiency (23 ±â€¯5 vs. 23 ±â€¯8%); release properties (15% released in 15 h in both cases); and apparent permeation across excised rat intestine (Papp, 8.8 ±â€¯0.8 vs. 10 ±â€¯1 cm/s). Then the differences in NP transport ratio through mucus (TR, 0.75 vs. 0.37) and adhesion to excised rat intestinal mucosa (adsorbed fraction, 23 ±â€¯3 vs. 45 ±â€¯2%) were ascribed to higher mucoadhesivity of NP QA-Ch-S-pro compared to NP QA-Ch. This directly influenced drug oral bioavailability in rats (Tmax, 1 vs. 2 h; AUC, 1.7 ±â€¯0.3 vs. 2.9 ±â€¯0.4 µg/mL min, for NP QA-Ch and NP QA-Ch-S-pro, respectively). Mucoadhesivity increases drug bioavailability by retaining NP at its absorption site and opposing its transit down the GI tract. Data on drug accumulation in rat liver allows the assertion that NP is absorbed by transcytosis across intestinal epithelium and transported from blood into liver by Kuppfer cells.


Asunto(s)
Dextranos/administración & dosificación , Sistemas de Liberación de Medicamentos , Fluoresceína-5-Isotiocianato/análogos & derivados , Nanopartículas , Polímeros/química , Adhesividad , Administración Oral , Animales , Área Bajo la Curva , Disponibilidad Biológica , Quitosano/química , Dextranos/química , Dextranos/farmacocinética , Portadores de Fármacos/química , Fluoresceína-5-Isotiocianato/administración & dosificación , Fluoresceína-5-Isotiocianato/química , Fluoresceína-5-Isotiocianato/farmacocinética , Ácido Hialurónico/química , Absorción Intestinal , Mucosa Intestinal/metabolismo , Masculino , Tamaño de la Partícula , Compuestos de Amonio Cuaternario/química , Ratas , Ratas Wistar , Compuestos de Sulfhidrilo/química , Porcinos
12.
J Nutr Sci ; 5: e23, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27293560

RESUMEN

The probiotic Lactobacillus gasseri SBT2055 (LG2055) has anti-obesity effects. Obesity is closely correlated with inflammation in adipose tissue, and maintaining adipose tissue in a less-inflamed state requires intestinal integrity or a barrier function to protect the intestine from the disruption that can be caused by a high-fat diet (HFD). Here, we examined the anti-inflammatory and intestinal barrier-protecting effects of LG2055 in C57BL/6 mice fed a normal-fat diet (NFD), HFD, or the HFD containing LG2055 (HFD-LG) for 21 weeks. HFD-LG intake significantly prevented HFD-induced increases in body weight, visceral fat mass, and the ratio of inflammatory-type macrophages to anti-inflammatory ones in adipose tissue. Mice fed the HFD showed higher intestinal permeability to a fluorescent dextran administered by oral administration and an elevated concentration of antibodies specific to lipopolysaccharides (LPS) in the blood compared with those fed the NFD, suggesting an increased penetration of the gut contents into the systemic circulation. These elevations of intestinal permeability and anti-LPS antibody levels were significantly suppressed in mice fed the HFD-LG. Moreover, treatment with LG2055 cells suppressed an increase in the cytokine-induced permeability of Caco-2 cell monolayers. These results suggest that LG2055 improves the intestinal integrity, reducing the entry of inflammatory substances like LPS from the intestine, which may lead to decreased inflammation in adipose tissue.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda