Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Lab Chip ; 24(5): 1266-1292, 2024 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-38226866

RESUMEN

The continued rise in metabolic diseases such as obesity and type 2 diabetes mellitus poses a global health burden, necessitating further research into factors implicated in the onset and progression of these diseases. Recently, the gut-immune axis, with diet as a main regulator, has been identified as a possible role player in their development. Translation of conventional 2D in vitro and animal models is however limited, while human studies are expensive and preclude individual mechanisms from being investigated. Lab-on-chip technology therefore offers an attractive new avenue to study gut-immune interactions. This review provides an overview of the influence of diet on gut-immune interactions in metabolic diseases and a critical analysis of the current state of lab-on-chip technology to study this axis. While there has been progress in the development of "immuno-competent" intestinal lab-on-chip models, with studies showing the ability of the technology to provide mechanical cues, support longer-term co-culture of microbiota and maintain in vivo-like oxygen gradients, platforms which combine all three and include intestinal and immune cells are still lacking. Further, immune cell types and inclusion of microenvironment conditions which enable in vivo-like immune cell dynamics as well as host-microbiome interactions are limited. Future model development should focus on combining these conditions to create an environment capable of hosting more complex microbiota and immune cells to allow further study into the effects of diet and related metabolites on the gut-immune ecosystem and their role in the prevention and development of metabolic diseases in humans.


Asunto(s)
Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Enfermedades Metabólicas , Animales , Humanos , Ecosistema , Enfermedades Metabólicas/metabolismo , Tecnología
2.
Mater Horiz ; 10(9): 3589-3600, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37318042

RESUMEN

Three-dimensional in vitro stem cell models have enabled a fundamental understanding of cues that direct stem cell fate. While sophisticated 3D tissues can be generated, technology that can accurately monitor these complex models in a high-throughput and non-invasive manner is not well adapted. Here we show the development of 3D bioelectronic devices based on the electroactive polymer poly(3,4-ethylenedioxythiophene)-poly(styrenesulfonate)-(PEDOT:PSS) and their use for non-invasive, electrical monitoring of stem cell growth. We show that the electrical, mechanical and wetting properties as well as the pore size/architecture of 3D PEDOT:PSS scaffolds can be fine-tuned simply by changing the processing crosslinker additive. We present a comprehensive characterization of both 2D PEDOT:PSS thin films of controlled thicknesses, and 3D porous PEDOT:PSS structures made by the freeze-drying technique. By slicing the bulky scaffolds we generate homogeneous, porous 250 µm thick PEDOT:PSS slices, constituting biocompatible 3D constructs able to support stem cell cultures. These multifunctional slices are attached on indium-tin oxide substrates (ITO) with the help of an electrically active adhesion layer, enabling 3D bioelectronic devices with a characteristic and reproducible, frequency dependent impedance response. This response changes drastically when human adipose derived stem cells (hADSCs) grow within the porous PEDOT:PSS network as revealed by fluorescence microscopy. The increase of cell population within the PEDOT:PSS porous network impedes the charge flow at the interface between PEDOT:PSS and ITO, enabling the interface resistance (R1) to be used as a figure of merit to monitor the proliferation of stem cells. The non-invasive monitoring of stem cell growth allows for the subsequent differentiation 3D stem cell cultures into neuron like cells, as verified by immunofluorescence and RT-qPCR measurements. The strategy of controlling important properties of 3D PEDOT:PSS structures simply by altering processing parameters can be applied for development of a number of stem cell in vitro models as well as stem cell differentiation pathways. We believe the results presented here will advance 3D bioelectronic technology for both fundamental understanding of in vitro stem cell cultures as well as the development of personalized therapies.


Asunto(s)
Células Madre Adultas , Electricidad , Humanos , Neuronas , Diferenciación Celular , Impedancia Eléctrica
3.
J Agric Food Chem ; 69(36): 10550-10561, 2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34460245

RESUMEN

Red wine is rich in phenolic compounds, which chiefly determine its characteristic taste. One of its major phenolic acid constituents for which an astringency, yet no clear contribution to bitter taste has been reported, is gallic acid (GA). In previous studies, we have demonstrated bitter-tasting constituents to regulate cellular proton secretion (PS) as a key mechanism of gastric acid secretion via activation of bitter taste sensing receptors (TAS2Rs). Here, we hypothesized a contributing role of GA to the red wine-stimulated effect on PS in human gastric tumor cells (HGT-1 cells). Sensory analyses revealed that 10 µM GA as the lowest concentration tested more bitter than tap water, with increasing bitter ratings up to 1000 µM. In HGT-1 cells, the concentration of 10 µM GA evoked the most pronounced effect on PS secretion, either when added to cells as in-water solution or when spiked to a red wine matrix. GA-spiking of Zweigelt and Blaufränkisch red wine samples up to a concentration of 10 µM resulted in an equally stimulated PS, whereas the non-GA-spiked wine samples demonstrated contrary effects on PS, indicating a functional role of GA on PS. Involvement of TAS2R4 in the GA-induced PS was verified by means of an HGT-1 homozygote CRISPR-Cas9 TAS2R4 knockout approach. Moreover, gene expression analyses revealed GA to increase TAS2R4. These results demonstrate a functional role of TAS2R4 in GA-evoked PS as a key mechanism of gastric acid secretion aiding digestion. Moreover, our data provide mechanistic insights, which will help to produce stomach-friendly red wines.


Asunto(s)
Gusto , Vino , Astringentes , Digestión , Ácido Gálico/farmacología , Ácido Gástrico , Humanos , Vino/análisis
4.
Int J Mol Sci ; 22(11)2021 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-34070942

RESUMEN

Among mammals, serotonin is predominantly found in the gastrointestinal tract, where it has been shown to participate in pathway-regulating satiation. For the stomach, vascular serotonin release induced by gastric distension is thought to chiefly contribute to satiation after food intake. However, little information is available on the capability of gastric cells to synthesize, release and respond to serotonin by functional changes of mechanisms regulating gastric acid secretion. We investigated whether human gastric cells are capable of serotonin synthesis and release. First, HGT-1 cells, derived from a human adenocarcinoma of the stomach, and human stomach specimens were immunostained positive for serotonin. In HGT-1 cells, incubation with the tryptophan hydroxylase inhibitor p-chlorophenylalanine reduced the mean serotonin-induced fluorescence signal intensity by 27%. Serotonin release of 147 ± 18%, compared to control HGT-1 cells (set to 100%) was demonstrated after treatment with 30 mM of the satiating amino acid L-Arg. Granisetron, a 5-HT3 receptor antagonist, reduced this L-Arg-induced serotonin release, as well as L-Arg-induced proton secretion. Similarly to the in vitro experiment, human antrum samples released serotonin upon incubation with 10 mM L-Arg. Overall, our data suggest that human parietal cells in culture, as well as from the gastric antrum, synthesize serotonin and release it after treatment with L-Arg via an HTR3-related mechanism. Moreover, we suggest not only gastric distension but also gastric acid secretion to result in peripheral serotonin release.


Asunto(s)
Arginina/farmacología , Ácido Gástrico/metabolismo , Células Parietales Gástricas/efectos de los fármacos , Protones , Serotonina/biosíntesis , Línea Celular Tumoral , Fenclonina/farmacología , Expresión Génica , Granisetrón/farmacología , Humanos , Concentración de Iones de Hidrógeno , Células Parietales Gástricas/citología , Células Parietales Gástricas/metabolismo , Inhibidores de Proteasas/farmacología , Receptores de Serotonina 5-HT3/genética , Receptores de Serotonina 5-HT3/metabolismo , Antagonistas de la Serotonina/farmacología , Estómago/citología , Estómago/efectos de los fármacos , Técnicas de Cultivo de Tejidos , Triptófano Hidroxilasa/antagonistas & inhibidores , Triptófano Hidroxilasa/genética , Triptófano Hidroxilasa/metabolismo
5.
J Agric Food Chem ; 68(11): 3434-3444, 2020 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-31891507

RESUMEN

This study aimed at identifying whether the bitter-tasting amino acids l-arginine (l-ARG) and l-isoleucine (l-ILE) differentially regulate mechanisms of gastric acid secretion in human parietal cells (HGT-1 cells) via activation of bitter taste sensing receptors (T2Rs). In a first set of experiments, involvement of T2Rs in l-ARG and l-ILE-modulated proton secretion was demonstrated by co-treatment of HGT-1 cells with T2R antagonists. Subsequent whole genome screenings by means of cDNA arrays revealed T2R1 as a prominent target for both amino acids. Next, the functional role of T2R1 was verified by means of a T2R1 CRISPR-Cas9 knock-out approach. Here, the effect of l-ARG on proton secretion decreased by 65.7 ± 21.9% and the effect of l-ILE increased by 93.2 ± 24.1% in HGT-1 T2R1 ko versus HGT-1 wt cells (p < 0.05). Overall, our results indicate differential effects of l-ARG and l-ILE on proton secretion in HGT-1 cells and our molecular docking studies predict distinct binding for these amino acids in the binding site of T2R1. Further studies will elucidate whether the mechanism of differential effects involves structure-specific ligand-biased signaling of T2R1 or additional cellular targets.


Asunto(s)
Isoleucina , Gusto , Aminoácidos , Arginina , Humanos , Simulación del Acoplamiento Molecular , Protones , Receptores Acoplados a Proteínas G/genética
6.
Mol Nutr Food Res ; 63(23): e1900133, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31535460

RESUMEN

SCOPE: Increasing the intake of satiety-enhancing food compounds represents a promising strategy for maintaining a healthy body weight. Recently, satiating effects for the capsaicinoid nonivamide have been demonstrated. As various proteins and amino acids have also been demonstrated to decrease energy intake, oral glucose tolerance test (oGTT)-based bolus interventions of 75 g glucose + 0.15 mg nonivamide (NV control) are tested with/without combination of a wheat protein hydrolysate (WPH: 2 g) and/or l-arginine (ARG: 3.2 g) for their satiating effects in 27 moderately overweight male subjects. METHODS AND RESULTS: Compared to NV control intervention, ARG and WPH + ARG treatment both reduce (p < 0.01) total calorie intake from a standardized breakfast by -5.9 ± 4.15% and -6.07 ± 4.38%, respectively. For the WPH + ARG intervention, increased mean plasma serotonin concentrations (AUC: 350 ± 218), quantitated by ELISA, and delayed gastric emptying, assessed by 13 C-Na-acetate breath test (-2.10 ± 0.51%, p < 0.05), are demonstrated compared to NV control. Correlation analysis between plasma serotonin and gastric emptying reveals a significant association after WPH ± ARG intervention (r = -0.396, p = 0.045). CONCLUSION: Combination of WPH and ARG enhances the satiating effect of nonivamide, providing opportunities to optimize satiating food formulations by low amounts of the individual food constituents.


Asunto(s)
Arginina/administración & dosificación , Capsaicina/análogos & derivados , Sobrepeso/psicología , Hidrolisados de Proteína/administración & dosificación , Saciedad/efectos de los fármacos , Triticum/química , Adulto , Capsaicina/farmacología , Estudios Cruzados , Ingestión de Energía , Vaciamiento Gástrico/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Serotonina/sangre , Método Simple Ciego
7.
J Agric Food Chem ; 66(26): 6762-6771, 2018 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-29879844

RESUMEN

Secretion of gastric acid, aimed at preventing bacterial growth and aiding the digestion of foods in the stomach, is chiefly stimulated by dietary intake of protein and amino acids (AAs). However, AAs' key structural determinants responsible for their effects on mechanisms regulating gastric acid secretion (GAS) have not been identified yet. In this study, AAs have been tested in the parietal cell model HGT-1 on GAS and on mRNA expression of genes regulating GAS. AAs' taste intensities from 0 (not bitter at all) to 10 (very bitter) were assessed in a sensory study, in which ARG (l: 6.42 ± 0.41; d: 4.62 ± 0.59) and ILE (l: 4.21 ± 0.43; d: 2.28 ± 0.33) were identified as bitter-tasting candidates in both isomeric forms. Pearson correlation showed that GAS in HGT-1 cells is directly associated with the bitter taste quality ( r: -0.654) in combination with the molecular weight of l-AA ( r: -0.685).


Asunto(s)
Aminoácidos/metabolismo , Ácido Gástrico/metabolismo , Células Parietales Gástricas/metabolismo , Gusto , Adulto , Aminoácidos/química , Línea Celular Tumoral , Humanos , Peso Molecular , Adulto Joven
8.
J Agric Food Chem ; 66(19): 4842-4852, 2018 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-29665689

RESUMEN

The noncaloric sweeteners (NCSs) cyclamate (Cycl) and acesulfame K (AceK) are widely added to foods and beverages. Little is known about their impact on gastric acid secretion (GAS), which is stimulated by dietary protein and bitter-tasting compounds. Since Cycl and AceK have a bitter off taste in addition to their sweet taste, we hypothesized they modulate mechanisms of GAS in human gastric parietal cells (HGT-1). HGT-1 cells were exposed to sweet tastants (50 mM of glucose, d-threonine, Cycl, or AceK) and analyzed for their intracellular pH index (IPX), as an indicator of proton secretion by means of a pH-sensitive dye, and for mRNA levels of GAS-associated genes by RT-qPCR. Since the NCSs act via the sweet taste-sensing receptor T1R2/T1R3, mRNA expression of the corresponding genes was analyzed in addition to immunocytochemical localization of the T1R2 and T1R3 receptor proteins. Exposure of HGT-1 cells to AceK or d-threonine increased the IPX to 0.60 ± 0.05 and 0.80 ± 0.04 ( P ≤ 0.05), respectively, thereby indicating a reduced secretion of protons, whereas Cycl demonstrated the opposite effect with IPX values of -0.69 ± 0.08 ( P ≤ 0.05) compared to controls (IPX = 0). Cotreatment with the T1R3-inhibitor lactisole as well as a TAS1R3 siRNA knock-down approach reduced the impact of Cycl, AceK, and d-thr on proton release ( P ≤ 0.05), whereas cotreatment with 10 mM glucose enhanced the NCS-induced effect ( P ≤ 0.05). Overall, we demonstrated Cycl and AceK as modulators of proton secretion in HGT-1 cells and identified T1R3 as a key element in this response.


Asunto(s)
Ciclamatos/metabolismo , Ácido Gástrico/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Edulcorantes/metabolismo , Tiazinas/metabolismo , Línea Celular Tumoral , Humanos , Receptores Acoplados a Proteínas G/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA