Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 575
Filtrar
1.
Cell Death Dis ; 15(4): 301, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38684650

RESUMEN

Understanding the mechanisms involved in colonic epithelial differentiation is key to unraveling the alterations causing inflammatory conditions and cancer. Organoid cultures provide an unique tool to address these questions but studies are scarce. We report a differentiation system toward enterocytes and goblet cells, the two major colonic epithelial cell lineages, using colon organoids generated from healthy tissue of colorectal cancer patients. Culture of these organoids in medium lacking stemness agents resulted in a modest ultrastructural differentiation phenotype with low-level expression of enterocyte (KLF4, KRT20, CA1, FABP2) and goblet cell (TFF2, TFF3, AGR2) lineage markers. BMP pathway activation through depletion of Noggin and addition of BMP4 resulted in enterocyte-biased differentiation. Contrarily, blockade of the Notch pathway using the γ-secretase inhibitor dibenzazepine (DBZ) favored goblet cell differentiation. Combination treatment with BMP4 and DBZ caused a balanced strong induction of both lineages. In contrast, colon tumor organoids responded poorly to BMP4 showing only weak signals of cell differentiation, and were unresponsive to DBZ. We also investigated the effects of 1α,25-dihydroxyvitamin D3 (calcitriol) on differentiation. Calcitriol attenuated the effects of BMP4 and DBZ on colon normal organoids, with reduced expression of differentiation genes and phenotype. Consistently, in normal organoids, calcitriol inhibited early signaling by BMP4 as assessed by reduction of the level of phospho-SMAD1/5/8. Our results show that BMP and Notch signaling play key roles in human colon stem cell differentiation to the enterocytic and goblet cell lineages and that calcitriol modulates these processes favoring stemness features.


Asunto(s)
Proteína Morfogenética Ósea 4 , Calcitriol , Proteínas Portadoras , Diferenciación Celular , Colon , Dibenzazepinas , Células Caliciformes , Factor 4 Similar a Kruppel , Organoides , Receptores Notch , Transducción de Señal , Humanos , Organoides/efectos de los fármacos , Organoides/metabolismo , Diferenciación Celular/efectos de los fármacos , Proteína Morfogenética Ósea 4/metabolismo , Colon/efectos de los fármacos , Colon/metabolismo , Colon/citología , Colon/patología , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Calcitriol/farmacología , Células Caliciformes/efectos de los fármacos , Células Caliciformes/metabolismo , Dibenzazepinas/farmacología , Linaje de la Célula/efectos de los fármacos , Enterocitos/metabolismo , Enterocitos/efectos de los fármacos , Enterocitos/citología , Vitamina D/farmacología
2.
Cells ; 11(20)2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36291149

RESUMEN

BACKGROUND: The intestinal lining renews itself in a programmed fashion that can be affected by adaptation to surgical procedures such as gastric bypass. METHODS: To assess adaptive mechanisms in the human intestine after Roux-en-Y gastric bypass (RYGB), we biopsied proximal jejunum at the anastomotic site during surgery to establish a baseline and endoscopically re-biopsied the same area 6-9 months after bypass for comparison. Laser microdissection was performed on pre- and post-RYGB biopsies to isolate enterocytes for RNA sequencing. RESULTS: RNA sequencing suggested significant decreases in gene expression associated with G2/M DNA damage checkpoint regulation of the cell cycle pathway, and significant increases in gene expression associated with the CDP-diacylglycerol biosynthesis pathway TCA cycle II pathway, and pyrimidine ribonucleotide salvage pathway after RYGB. Since Schlafen 12 (SLFN12) is reported to influence enterocytic differentiation, we stained mucosa for SLFN12 and observed increased SLFN12 immunoreactivity. We investigated SLFN12 overexpression in HIEC-6 and FHs 74 Int intestinal epithelial cells and observed similar increased expression of the following genes that were also increased after RYGB: HES2, CARD9, SLC19A2, FBXW7, STXBP4, SPARCL1, and UTS. CONCLUSIONS: Our data suggest that RYGB promotes SLFN12 protein expression, cellular mechanism and replication pathways, and genes associated with differentiation and restitution (HES2, CARD9, SLC19A2), as well as obesity-related genes (FBXW7, STXBP4, SPARCL1, UTS).


Asunto(s)
Diferenciación Celular , Enterocitos , Derivación Gástrica , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular , Obesidad , Humanos , Citidina Difosfato Diglicéridos/metabolismo , Enterocitos/citología , Enterocitos/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Expresión Génica , Intestinos , Proteínas de Transporte de Membrana/metabolismo , Obesidad/genética , Obesidad/cirugía , Obesidad/metabolismo , Análisis de Secuencia de ARN , Proteínas de Transporte Vesicular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Diferenciación Celular/genética
3.
Biochim Biophys Acta Biomembr ; 1864(4): 183868, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35063401

RESUMEN

Iron is a key element in cell function; however, its excess in iron overload conditions can be harmful through the generation of reactive oxygen species (ROS) and cell oxidative stress. Activity of Na,K-ATPase has been shown to be implicated in cellular iron uptake and iron modulates the Na,K-ATPase function from different tissues. In this study, we determined the effect of iron overload on Na,K-ATPase activity and established the role that isoforms and conformational states of this enzyme has on this effect. Total blood and membrane preparations from erythrocytes (ghost cells), as well as pig kidney and rat brain cortex, and enterocytes cells (Caco-2) were used. In E1-related subconformations, an enzyme activation effect by iron was observed, and in the E2-related subconformations enzyme inhibition was observed. The enzyme's kinetic parameters were significantly changed only in the Na+ curve in ghost cells. In contrast to Na,K-ATPase α2 and α3 isoforms, activation was not observed for the α1 isoform. In Caco-2 cells, which only contain Na,K-ATPase α1 isoform, the FeCl3 increased the intracellular storage of iron, catalase activity, the production of H2O2 and the expression levels of the α1 isoform. In contrast, iron did not affect lipid peroxidation, GSH content, superoxide dismutase and Na,K-ATPase activities. These results suggest that iron itself modulates Na,K-ATPase and that one or more E1-related subconformations seems to be determinant for the sensitivity of iron modulation through a mechanism in which the involvement of the Na, K-ATPase α3 isoform needs to be further investigated.


Asunto(s)
Adenosina Trifosfato/metabolismo , Cloruros/química , Compuestos Férricos/química , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Células CACO-2 , Cloruros/metabolismo , Enterocitos/citología , Enterocitos/metabolismo , Membrana Eritrocítica/química , Membrana Eritrocítica/metabolismo , Compuestos Férricos/metabolismo , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Ratas , ATPasa Intercambiadora de Sodio-Potasio/genética , Porcinos
4.
Biomolecules ; 11(12)2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34944413

RESUMEN

Atherosclerosis is the main cause of cardiovascular diseases with high prevalence worldwide. A promising therapeutic strategy to reverse atherosclerotic process is to improve the athero-protective potential of high-density lipoproteins (HDL). Since the small intestine is a source of HDL, we aimed to activate transcription of the endogenous HDL major proteins, apolipoprotein AI (ApoAI) and paraoxonase 1 (PON1), in enterocytes, and to evaluate their potential to correct the pro-inflammatory status of endothelial cells (EC). Caco-2 enterocytes were transfected with CRISPR activation plasmids targeting ApoAI or PON1, and their gene and protein expression were measured in cells and conditioned medium (CM). ATP binding cassette A1 and G8 transporters (ABCA1, ABCG8), scavenger receptor BI (SR-BI), and transcription regulators peroxisome proliferator-activated receptor γ (PPARγ), liver X receptors (LXRs), and sirtuin-1 (SIRT1) were assessed. Anti-inflammatory effects of CM from transfected enterocytes were estimated through its ability to inhibit tumor necrosis factor α (TNFα) activation of EC. Transcriptional activation of ApoAI or PON1 in enterocytes induces: (i) increase of their gene and protein expression, and secretion in CM; (ii) stimulation of ABCA1/G8 and SR-BI; (iii) upregulation of PPARγ, LXRs, and SIRT1. CM from transfected enterocytes attenuated the TNFα-induced inflammatory and oxidative stress in EC, by decreasing TNF receptor 1, monocyte chemoattractant protein-1, and p22phox. In conclusion, transcriptional activation of endogenous ApoAI or PON1 in enterocytes by CRISPR/dCas9 system is a realistic approach to stimulate biogenesis and function of major HDL proteins which can regulate cholesterol efflux transporters and reduce the inflammatory stress in activated EC.


Asunto(s)
Apolipoproteína A-I/genética , Arildialquilfosfatasa/genética , Células Endoteliales/citología , Enterocitos/citología , Apolipoproteína A-I/metabolismo , Arildialquilfosfatasa/metabolismo , Sistemas CRISPR-Cas , Células CACO-2 , Medios de Cultivo Condicionados/química , Células Endoteliales/metabolismo , Enterocitos/metabolismo , Regulación de la Expresión Génica , Humanos , Lipoproteínas HDL/metabolismo , Estrés Oxidativo , Activación Transcripcional , Factor de Necrosis Tumoral alfa/metabolismo
5.
Elife ; 102021 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-34553686

RESUMEN

The gut is the primary interface between an animal and food, but how it adapts to qualitative dietary variation is poorly defined. We find that the Drosophila midgut plastically resizes following changes in dietary composition. A panel of nutrients collectively promote gut growth, which sugar opposes. Diet influences absolute and relative levels of enterocyte loss and stem cell proliferation, which together determine cell numbers. Diet also influences enterocyte size. A high sugar diet inhibits translation and uncouples intestinal stem cell proliferation from expression of niche-derived signals, but, surprisingly, rescuing these effects genetically was not sufficient to modify diet's impact on midgut size. However, when stem cell proliferation was deficient, diet's impact on enterocyte size was enhanced, and reducing enterocyte-autonomous TOR signaling was sufficient to attenuate diet-dependent midgut resizing. These data clarify the complex relationships between nutrition, epithelial dynamics, and cell size, and reveal a new mode of plastic, diet-dependent organ resizing.


Asunto(s)
Dieta , Drosophila melanogaster/crecimiento & desarrollo , Tracto Gastrointestinal/crecimiento & desarrollo , Animales , Animales Modificados Genéticamente , Proliferación Celular , Drosophila melanogaster/fisiología , Enterocitos/citología , Femenino , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/fisiología , Masculino , Nicho de Células Madre
6.
Cell Host Microbe ; 29(9): 1407-1420.e5, 2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34348092

RESUMEN

The parasite Cryptosporidium invades and replicates in intestinal epithelial cells and is a leading cause of diarrheal disease and early childhood mortality. The molecular mechanisms that underlie infection and pathogenesis are largely unknown. Here, we delineate the events of host cell invasion and uncover a mechanism unique to Cryptosporidium. We developed a screen to identify parasite effectors, finding the injection of multiple parasite proteins into the host from the rhoptry organelle. These factors are targeted to diverse locations within the host cell and its interface with the parasite. One identified effector, rhoptry protein 1 (ROP1), accumulates in the terminal web of enterocytes through direct interaction with the host protein LIM domain only 7 (LMO7) an organizer of epithelial cell polarity and cell-cell adhesion. Genetic ablation of LMO7 or ROP1 in mice or parasites, respectively, impacts parasite burden in vivo in opposite ways. Taken together, these data provide molecular insight into how Cryptosporidium manipulates its intestinal host niche.


Asunto(s)
Criptosporidiosis/patología , Cryptosporidium parvum/patogenicidad , Enterocitos/parasitología , Proteínas con Dominio LIM/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Protozoarias/metabolismo , Factores de Transcripción/metabolismo , Animales , Células CACO-2 , Adhesión Celular/fisiología , Línea Celular , Modelos Animales de Enfermedad , Enterocitos/citología , Células Epiteliales/parasitología , Células HEK293 , Interacciones Huésped-Parásitos/fisiología , Humanos , Proteínas con Dominio LIM/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Orgánulos/metabolismo , Factores de Transcripción/genética
7.
Cells ; 10(6)2021 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-34205481

RESUMEN

We derived two novel cell lines from rainbow trout (RT) proximal (RTpi-MI) and distal intestine (RTdi-MI) and compared them with the previously established continuous cell line RTgutGC. Intestinal stem cells, differentiating and differentiated epithelial cells, and connective cells were found in all cell lines. The cell lines formed a polarized barrier, which was not permeable to large molecules and absorbed proline and glucose. High seeding density induced their differentiation into more mature phenotypes, as indicated by the downregulation of intestinal stem cell-related genes (i.e., sox9, hopx and lgr5), whereas alkaline phosphatase activity was upregulated. Other enterocyte markers (i.e., sglt1 and pept1), however, were not regulated as expected. In all cell lines, the presence of a mixed population of epithelial and stromal cells was characterized for the first time. The expression by the stromal component of lgr5, a stem cell niche regulatory molecule, may explain why these lines proliferate stably in vitro. Although most parameters were conserved among the three cell lines, some significant differences were observed, suggesting that characteristics typical of each tract are partly conserved in vitro as well.


Asunto(s)
Enterocitos , Oncorhynchus mykiss/metabolismo , Animales , Línea Celular/citología , Línea Celular/metabolismo , Enterocitos/citología , Enterocitos/metabolismo
8.
Am J Physiol Cell Physiol ; 321(3): C471-C488, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34288721

RESUMEN

Intestinal NaCl, HCO3-, and fluid absorption are strongly dependent on apical Na+/H+ exchange. The intestine expresses three presumably apical sodium-hydrogen exchanger (NHE) isoforms: NHE2, NHE3, and NHE8. We addressed the role of NHE8 [solute carrier 9A8 (SLC9A8)] and its interplay with NHE2 (SLC9A2) in luminal proton extrusion during acute and chronic enterocyte acidosis and studied the differential effects of NHE8 and NHE2 on enterocyte proliferation. In contrast to NHE3, which was upregulated in differentiated versus undifferentiated colonoids, the expression of NHE2 and NHE8 remained constant during differentiation of colonoids and Caco2Bbe cells. Heterogeneously expressed Flag-tagged rat (r)Nhe8 and human (h)NHE8 translocated to the apical membrane of Caco2Bbe cells. rNhe8 and hNHE8, when expressed in NHE-deficient PS120 fibroblasts showed higher sensitivity to HOE642 compared to NHE2. Lentiviral shRNA knockdown of endogenous NHE2 in Caco2Bbe cells (C2Bbe/shNHE2) resulted in a decreased steady-state intracellular pH (pHi), an increased NHE8 mRNA expression, and augmented NHE8-mediated apical NHE activity. Lentiviral shRNA knockdown of endogenous NHE8 in Caco2Bbe cells (C2Bbe/shNHE8) resulted in a decreased steady-state pHi as well, accompanied by decreased NHE2 mRNA expression and activity, which together contributed to reduced apical NHE activity in the NHE8-knockdown cells. Chronic acidosis increased NHE8 but not NHE2 mRNA expression. Alterations in NHE2 and NHE8 expression/activity affected proliferation, with C2Bbe/shNHE2 cells having lower and C2Bbe/shNHE8 having higher proliferative capacity, accompanied by amplified ERK1/2 signaling pathway and increased EGFR expression in the latter cell line. Thus, both Na+/H+ exchangers have distinct functions during cellular homeostasis by triggering different signaling pathways to regulate cellular proliferation and pHi control.


Asunto(s)
Colon/metabolismo , Enterocitos/metabolismo , Intercambiadores de Sodio-Hidrógeno/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Colon/citología , Colon/efectos de los fármacos , Enterocitos/citología , Enterocitos/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Guanidinas/farmacología , Células HT29 , Homeostasis/genética , Humanos , Concentración de Iones de Hidrógeno , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Organoides/citología , Organoides/efectos de los fármacos , Organoides/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Transducción de Señal , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Intercambiadores de Sodio-Hidrógeno/metabolismo , Sulfonas/farmacología
9.
Cells ; 10(6)2021 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-34072441

RESUMEN

Paneth cell defects in Crohn's disease (CD) patients (called the Type I phenotype) are associated with worse clinical outcomes. Recent studies have implicated mitochondrial dysfunction in Paneth cells as a mediator of ileitis in mice. We hypothesized that CD Paneth cells exhibit impaired mitochondrial health and that mitochondrial-targeted therapeutics may provide a novel strategy for ileal CD. Terminal ileal mucosal biopsies from adult CD and non-IBD patients were characterized for Paneth cell phenotyping and mitochondrial damage. To demonstrate the response of mitochondrial-targeted therapeutics in CD, biopsies were treated with vehicle or Mito-Tempo, a mitochondrial-targeted antioxidant, and RNA transcriptome was analyzed. During active CD inflammation, the epithelium exhibited mitochondrial damage evident in Paneth cells, goblet cells, and enterocytes. Independent of inflammation, Paneth cells in Type I CD patients exhibited mitochondrial damage. Mito-Tempo normalized the expression of interleukin (IL)-17/IL-23, lipid metabolism, and apoptotic gene signatures in CD patients to non-IBD levels. When stratified by Paneth cell phenotype, the global tissue response to Mito-Tempo in Type I patients was associated with innate immune, lipid metabolism, and G protein-coupled receptor (GPCR) gene signatures. Targeting impaired mitochondria as an underlying contributor to inflammation provides a novel treatment approach for CD.


Asunto(s)
Antioxidantes/uso terapéutico , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/metabolismo , Inflamación/tratamiento farmacológico , Mitocondrias/metabolismo , Biopsia/métodos , Enterocitos/citología , Epitelio/efectos de los fármacos , Epitelio/patología , Humanos , Metabolismo de los Lípidos/fisiología , Células de Paneth/patología , Fenotipo
10.
Cell Mol Gastroenterol Hepatol ; 12(3): 943-981, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34102314

RESUMEN

BACKGROUND & AIMS: The use of antibiotics (ABs) is a common practice during the first months of life. ABs can perturb the intestinal microbiota, indirectly influencing the intestinal epithelial cells (IECs), but can also directly affect IECs independent of the microbiota. Previous studies have focused mostly on the impact of AB treatment during adulthood. However, the difference between the adult and neonatal intestine warrants careful investigation of AB effects in early life. METHODS: Neonatal mice were treated with a combination of amoxicillin, vancomycin, and metronidazole from postnatal day 10 to 20. Intestinal permeability and whole-intestine gene and protein expression were analyzed. IECs were sorted by a fluorescence-activated cell sorter and their genome-wide gene expression was analyzed. Mouse fetal intestinal organoids were treated with the same AB combination and their gene and protein expression and metabolic capacity were determined. RESULTS: We found that in vivo treatment of neonatal mice led to decreased intestinal permeability and a reduced number of specialized vacuolated cells, characteristic of the neonatal period and necessary for absorption of milk macromolecules. In addition, the expression of genes typically present in the neonatal intestinal epithelium was lower, whereas the adult gene expression signature was higher. Moreover, we found altered epithelial defense and transepithelial-sensing capacity. In vitro treatment of intestinal fetal organoids with AB showed that part of the consequences observed in vivo is a result of the direct action of the ABs on IECs. Lastly, ABs reduced the metabolic capacity of intestinal fetal organoids. CONCLUSIONS: Our results show that early life AB treatment induces direct and indirect effects on IECs, influencing their maturation and functioning.


Asunto(s)
Amoxicilina/administración & dosificación , Antibacterianos/administración & dosificación , Redes Reguladoras de Genes/efectos de los fármacos , Intestinos/metabolismo , Metronidazol/administración & dosificación , Vancomicina/administración & dosificación , Amoxicilina/efectos adversos , Animales , Animales Recién Nacidos , Antibacterianos/efectos adversos , Modelos Animales de Enfermedad , Enterocitos/citología , Enterocitos/efectos de los fármacos , Enterocitos/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Intestinos/citología , Intestinos/efectos de los fármacos , Metronidazol/efectos adversos , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Permeabilidad/efectos de los fármacos , Atención Posnatal , Vacuolas/efectos de los fármacos , Vacuolas/metabolismo , Vancomicina/efectos adversos
11.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34016750

RESUMEN

Adult organisms must sense and adapt to environmental fluctuations. In high-turnover tissues such as the intestine, these adaptive responses require rapid changes in gene expression that, in turn, likely involve posttranscriptional gene control. However, intestinal-tissue-specific microRNA (miRNA)-mediated regulatory pathways remain unexplored. Here, we report the role of an intestinal-specific miRNA, miR-958, that non-cell autonomously regulates stem cell numbers during tissue homeostasis and regeneration in the Drosophila adult midgut. We identify its downstream target cabut, the Drosophila ortholog of mammalian KLF10/11 transcription factors, which mediates this miR-958 function by promoting paracrine enterocyte-to-stem-cell bone morphogenetic protein (BMP) signaling. We also show that mature miR-958 levels transiently decrease in response to stress and that this decrease is required for proper stem cell expansion during tissue regeneration. In summary, we have identified a posttranscriptional mechanism that modulates BMP signaling activity within Drosophila adult intestinal tissue during both normal homeostasis and tissue regeneration to regulate intestinal stem cell numbers.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Enterocitos/metabolismo , MicroARNs/genética , Células Madre/metabolismo , Factores de Transcripción/genética , Animales , Bleomicina/farmacología , Proteínas Morfogenéticas Óseas/metabolismo , Recuento de Células , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Enterocitos/citología , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Homeostasis/genética , MicroARNs/metabolismo , Regeneración/genética , Transducción de Señal , Células Madre/citología , Factores de Transcripción/metabolismo
12.
Cells ; 10(4)2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33917333

RESUMEN

The endoderm, differentiated from human induced pluripotent stem cells (iPSCs), can differentiate into the small intestine and liver, which are vital for drug absorption and metabolism. The development of human iPSC-derived enterocytes (HiEnts) and hepatocytes (HiHeps) has been reported. However, pharmacokinetic function-deficiency of these cells remains to be elucidated. Here, we aimed to develop an efficient differentiation method to induce endoderm formation from human iPSCs. Cells treated with activin A for 168 h expressed higher levels of endodermal genes than those treated for 72 h. Using activin A (days 0-7), CHIR99021 and PI-103 (days 0-2), and FGF2 (days 3-7), the hiPSC-derived endoderm (HiEnd) showed 97.97% CD-117 and CD-184 double-positive cells. Moreover, HiEnts derived from the human iPSC line Windy had similar or higher expression of small intestine-specific genes than adult human small intestine. Activities of the drug transporter P-glycoprotein and drug-metabolizing enzyme cytochrome P450 (CYP) 3A4/5 were confirmed. Additionally, Windy-derived HiHeps expressed higher levels of hepatocyte- and pharmacokinetics-related genes and proteins and showed higher CYP3A4/5 activity than those derived through the conventional differentiation method. Thus, using this novel method, the differentiated HiEnts and HiHeps with pharmacokinetic functions could be used for drug development.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Endodermo/citología , Enterocitos/citología , Hepatocitos/citología , Células Madre Pluripotentes Inducidas/citología , Activinas/farmacología , Proteína Morfogenética Ósea 4/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Cultivadas , Dimetilsulfóxido/farmacología , Enterocitos/efectos de los fármacos , Células Nutrientes/citología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Furanos/farmacología , Hepatocitos/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Intestino Delgado/citología , Línea Primitiva/citología , Piridinas/farmacología , Pirimidinas/farmacología , Reproducibilidad de los Resultados
13.
Mucosal Immunol ; 14(5): 1017-1037, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33859369

RESUMEN

The mechanisms that maintain intestinal homeostasis despite constant exposure of the gut surface to multiple environmental antigens and to billions of microbes have been scrutinized over the past 20 years with the goals to gain basic knowledge, but also to elucidate the pathogenesis of inflammatory bowel diseases (IBD) and to identify therapeutic targets for these severe diseases. Considerable insight has been obtained from studies based on gene inactivation in mice as well as from genome wide screens for genetic variants predisposing to human IBD. These studies are, however, not sufficient to delineate which pathways play key nonredundant role in the human intestinal barrier and to hierarchize their respective contribution. Here, we intend to illustrate how such insight can be derived from the study of human Mendelian diseases, in which severe intestinal pathology results from single gene defects that impair epithelial and or hematopoietic immune cell functions. We suggest that these diseases offer the unique opportunity to study in depth the pathogenic mechanisms leading to perturbation of intestinal homeostasis in humans. Furthermore, molecular dissection of monogenic intestinal diseases highlights key pathways that might be druggable and therapeutically targeted in common forms of IBD.


Asunto(s)
Interacciones Microbiota-Huesped/inmunología , Inmunidad Mucosa , Inmunomodulación , Mucosa Intestinal/fisiología , Inmunidad Adaptativa , Animales , Transporte Biológico , Biomarcadores , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Susceptibilidad a Enfermedades , Enterocitos/citología , Enterocitos/inmunología , Enterocitos/metabolismo , Regulación de la Expresión Génica , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/inmunología , Predisposición Genética a la Enfermedad , Hematopoyesis/genética , Hematopoyesis/inmunología , Homeostasis , Interacciones Microbiota-Huesped/genética , Humanos , Inmunidad Innata , Mutación , Transducción de Señal
14.
Int J Mol Sci ; 22(7)2021 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-33916868

RESUMEN

MicroRNAs (miRNAs) are small RNAs present in extracellular vesicles (EVs) that, when transferred to a target cell, affect its biological functions. Plant miRNAs regulate the expression of certain mammalian genes. Here, we characterized EVs in fruit and vegetable juice, and their miRNA cargo, and investigated whether such miRNA-containing EVs could be taken up by mammalian enterocytes in vitro. Using filtration and ultra-centrifugation methods, EVs were purified from commercially available and manually squeezed plant juice. EV morphological features and subcellular localization were analyzed using the NanoSight tracking system and electron microscopy. Plant EV miRNA levels were evaluated using quantitative reverse transcription PCR. For the in vitro EV uptake experiments, rat intestinal epithelial cells (IEC6) were used. Plant EVs shared morphological features with mammalian EVs and contained miR156a-5p, miR166a-3p, and miR168a-5p. EVs were present in the cell sap-filled central vacuoles and were taken up by IEC6 cells. Edible plant cells produce EVs that contain various miRNAs and release them into the central vacuole. The exogenous plant EVs are taken up by mammalian enterocytes in vitro. These findings suggest the possibility that exogenous plant miRNAs carried by EVs can be absorbed via the gastrointestinal tract.


Asunto(s)
Enterocitos/metabolismo , Exosomas/química , Jugos de Frutas y Vegetales , MicroARNs , Nanopartículas , Animales , Línea Celular , Enterocitos/citología , MicroARNs/farmacocinética , MicroARNs/farmacología , Nanopartículas/química , Nanopartículas/uso terapéutico , Ratas
15.
Nutrients ; 13(3)2021 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-33801889

RESUMEN

Background: The aim of this study was to examine the anti-inflammatory and anti-apoptotic patterns of omega-3 polyunsaturated fatty acids (n-3 PUFAs) during methotrexate (MTX) induced intestinal damage in cell culture and in a rat model. Methods: Non-treated and treated with MTX HT 29 and HCT116cells were exposed to increasing doses of n-3 PUFAs and cell viability was evaluated using PrestoBlue® assay. Male Sprague-Dawley rats were divided into 4 experimental groups: Control rats, CONTR+n-3 PUFA rats that were treated with oral n-3 PUFA, MTX rats were treated with MTX given IP, and MTX+n-3 PUFA rats were treated with oral n-3 PUFA before and following injection of MTX. Intestinal mucosal parameters and mucosal inflammation, enterocyte proliferation and apoptosis, TNF-α in mucosal tissue and plasma (ELISA), NF-κB, COX-2, TNF-α, Fas, FasL, Fadd, Bid, Bax and Bcl-2gene and protein levels were determined 72 h following MTX injection. Results: Exposure of HT 29 and HCT116cells to n-3 PUFA attenuated inhibiting effects of MTX on cell viability. MTX-n-3 PUFA rats demonstrated a lower intestinal injury score and enhanced intestinal repair. A significant decrease in enterocyte apoptosis in MTX+n-3 PUFA rats was accompanied by decreased TNF-α, FAS, FasL, FADD and BID mRNA levels. Decreased NF-κB, COX-2 and TNF-α levels in mucosa was accompanied by a decreased number of IELs and macrophages. Conclusions: n-3 PUFAs inhibit NF-κB/COX-2 induced production of pro-inflammatory cytokines and inhibit cell apoptosis mainly by extrinsic pathway in rats with MTX-induced intestinal damage.


Asunto(s)
Antiinflamatorios/administración & dosificación , Apoptosis/efectos de los fármacos , Citocinas/metabolismo , Ácidos Grasos Omega-3/administración & dosificación , Mucosa Intestinal/citología , Metotrexato/toxicidad , Mucositis/terapia , Animales , Antiinflamatorios/farmacología , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Enterocitos/citología , Enterocitos/efectos de los fármacos , Ácidos Grasos Omega-3/farmacología , Células HCT116 , Células HT29 , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Masculino , Mucositis/inducido químicamente , Mucositis/metabolismo , Mucositis/patología , FN-kappa B/metabolismo , Ratas , Ratas Sprague-Dawley
16.
Genetics ; 218(1)2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-33693718

RESUMEN

The molecular mechanisms by which stem cell proliferation is precisely controlled during the course of regeneration are poorly understood. Namely, how a damaged tissue senses when to terminate the regeneration process, inactivates stem cell mitotic activity, and organizes ECM integrity remain fundamental unanswered questions. The Drosophila midgut intestinal stem cell (ISC) offers an excellent model system to study the molecular basis for stem cell inactivation. Here, we show that a novel gene, CG6967 or dMOV10, is induced at the termination stage of midgut regeneration, and shows an inhibitory effect on ISC proliferation. dMOV10 encodes a putative component of the microRNA (miRNA) gene silencing complex (miRISC). Our data, along with previous studies on the mammalian MOV10, suggest that dMOV10 is not a core member of miRISC, but modulates miRISC activity as an additional component. Further analyses identified direct target mRNAs of dMOV10-containing miRISC, including Daughter against Dpp (Dad), a known inhibitor of BMP/TGF-ß signaling. We show that RNAi knockdown of Dad significantly impaired ISC division during regeneration. We also identified six miRNAs that are induced at the termination stage and their potential target transcripts. One of these miRNAs, mir-1, is required for proper termination of ISC division at the end of regeneration. We propose that miRNA-mediated gene regulation contributes to the precise control of Drosophila midgut regeneration.


Asunto(s)
Drosophila/fisiología , Mucosa Intestinal/citología , Regeneración/fisiología , Células Madre/citología , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Autorrenovación de las Células/genética , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Enterocitos/citología , Intestinos/citología , MicroARNs/genética , MicroARNs/metabolismo , ARN Helicasas , Complejo Silenciador Inducido por ARN/genética , Complejo Silenciador Inducido por ARN/metabolismo , Transducción de Señal , Células Madre/metabolismo
17.
BMC Mol Cell Biol ; 22(1): 12, 2021 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-33579204

RESUMEN

BACKGROUND: Enterocytes exert an absorptive and protective function in the intestine, and they encounter many different challenging factors such as feed, bacteria, and parasites. An intestinal epithelial in vitro model can help to understand how enterocytes are affected by these factors and contribute to the development of strategies against pathogens. RESULTS: The present study describes a novel method to culture and maintain primary chicken enterocytes and their characterization by immunofluorescence and biomolecular approaches. Starting from 19-day-old chicken embryos it was possible to isolate viable intestinal cell aggregates that can expand and produce a self-maintaining intestinal epithelial cell population that survives until 12 days in culture. These cells resulted positive in immunofluorescence to Cytokeratin 18, Zonula occludens 1, Villin, and Occludin that are common intestinal epithelial markers, and negative to Vimentin that is expressed by endothelial cells. Cells were cultured also on Transwell® permeable supports and trans-epithelial electrical resistance, was measured. This value gradually increased reaching 64 Ω*cm2 7 days after seeding and it remained stable until day 12. CONCLUSIONS: Based on these results it was confirmed that it is possible to isolate and maintain chicken intestinal epithelial cells in culture and that they can be suitable as in vitro intestinal model for further studies.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Separación Celular/métodos , Enterocitos/citología , Mucosa Intestinal/citología , Mucosa Intestinal/embriología , Animales , Proliferación Celular , Células Cultivadas , Embrión de Pollo , Pollos , Colagenasas/metabolismo , Medios de Cultivo , Desarrollo Embrionario , Mucosa Intestinal/enzimología , Tripsina/farmacología
18.
Stem Cell Reports ; 16(2): 295-308, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33513361

RESUMEN

We aimed to establish an in vitro differentiation procedure to generate matured small intestinal cells mimicking human small intestine from human-induced pluripotent stem cells (iPSCs). We previously reported the efficient generation of CDX2-expressing intestinal progenitor cells from embryonic stem cells (ESCs) using 6-bromoindirubin-3'-oxime (BIO) and (3,5-difluorophenylacetyl)-L-alanyl-L-2-phenylglycine tert-butyl ester (DAPT) to treat definitive endodermal cells. Here, we demonstrate the generation of enterocyte-like cells by culturing human iPSC-derived intestinal progenitor cells on a collagen vitrigel membrane (CVM) and treating cells with a simple maturation medium containing BIO, DMSO, dexamethasone, and activated vitamin D3. Functional tests further confirmed that these iPSC-derived enterocyte-like cells exhibit P-gp- and BCRP-mediated efflux and cytochrome P450 3A4 (CYP3A4)-mediated metabolism. We concluded that hiPS cell-derived enterocyte-like cells can be used as a model for the evaluation of drug transport and metabolism studies in the human small intestine.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Enterocitos/citología , Enterocitos/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Intestino Delgado/citología , Intestino Delgado/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Adulto , Diferenciación Celular , Línea Celular , Células Cultivadas , Colágeno/metabolismo , Medios de Cultivo , Citocromo P-450 CYP3A/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Adulto Joven
19.
J Fish Biol ; 98(5): 1349-1362, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33410520

RESUMEN

We report the effects of food deprivation on the early development of Pacific red snapper Lutjanus peru during the first days of development. The point of no return (PNR) was determined using the feeding incidence after a delay in first feeding. The gradual deterioration of the larvae during food deprivation was recorded using morphometric, histological, enzymatic and biochemical analysis. The time to reach the PNR was 120 h after hatching. Morphologically, the total length, muscle height, head length, tail length and pectoral angle showed the biggest reductions and their growth coefficients changed significantly during food deprivation. Histologically, enterocyte height also was reduced significantly. The protein concentration and activities of the digestive enzymes trypsin, cathepsin-like and lipase showed a significant decrease; meanwhile, amylase activity remained constant during food deprivation. The concentration of total essential free amino acids (EFAAs) decreased significantly while that of the nonessential free amino acids (NEFAAs) remain stable during food deprivation. The most abundant EFAAs were lysine, leucine, isoleucine and valine; the most abundant NEFAAs were alanine, glycine and glutamate, suggesting a more prominent role as energy substrates. At the time of the PNR the concentration of almost all the free amino acids showed a significant decrease. Early food deprivation has a significant impact on the morphology and biochemical characteristics of L. peru. These results suggest that initial feeding of L. peru should begin within 3 days of yolk sac depletion to avoid the PNR. Further studies are necessary to confirm and validate the characters identified in this study as biomarkers of starvation under culture conditions and evaluate their possible utility in ichthyoplankton surveys.


Asunto(s)
Enterocitos/citología , Privación de Alimentos , Músculos/citología , Perciformes/anatomía & histología , Perciformes/crecimiento & desarrollo , Aminoácidos/sangre , Animales , Enterocitos/patología , Músculos/química , Músculos/patología , Perciformes/metabolismo
20.
Am J Physiol Gastrointest Liver Physiol ; 320(3): G258-G271, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33074011

RESUMEN

There is increasing evidence that the study of normal human enteroids duplicates many known aspects of human intestinal physiology. However, this epithelial cell-only model lacks the many nonepithelial intestinal cells present in the gastrointestinal tract and exposure to the mechanical forces to which the intestine is exposed. We tested the hypothesis that physical shear forces produced by luminal and blood flow would provide an intestinal model more closely resembling normal human jejunum. Jejunal enteroid monolayers were studied in the Emulate, Inc. Intestine-Chip under conditions of constant luminal and basolateral flow that was designed to mimic normal intestinal fluid flow, with human umbilical vein endothelial cells (HUVECs) on the basolateral surface and with Wnt3A, R-spondin, and Noggin only on the luminal surface. The jejunal enteroids formed monolayers that remained confluent for 6-8 days, began differentiating at least as early as day 2 post plating, and demonstrated continuing differentiation over the entire time of the study, as shown by quantitative real-time polymerase chain reaction and Western blot analysis. Differentiation impacted villus genes and proteins differently with early expression of regenerating family member 1α (REG1A), early reduction to a low but constant level of expression of Na+-K+-2Cl- cotransporter 1 (NKCC1), and increasing expression of sucrase-isomaltase (SI) and downregulated in adenoma (DRA). These results were consistent with continual differentiation, as was shown to occur in mouse villus enterocytes. Compared with differentiated enteroid monolayers grown on Transwell inserts, enteroids exposed to flow were more differentiated but exhibited increased apoptosis and reduced carbohydrate metabolism, as shown by proteomic analysis. This study of human jejunal enteroids-on-chip suggests that luminal and basolateral flow produce a model of continual differentiation over time and NaCl absorption that mimics normal intestine and should provide new insights in intestinal physiology.NEW & NOTEWORTHY This study showed that polarized enteroid models in which there is no basolateral Wnt3a, are differentiated, regardless of the Wnt3a status of the apical media. The study supports the concept that in the human intestine villus differentiation is not an all or none phenomenon, demonstrating that at different days after lack of basolateral Wnt exposure, clusters of genes and proteins exist geographically along the villus with different domains having different functions.


Asunto(s)
Diferenciación Celular , Yeyuno/citología , Microfluídica/métodos , Cultivo Primario de Células/métodos , Estrés Mecánico , Adulto , Apoptosis , Proteínas Portadoras/metabolismo , Células Cultivadas , Enterocitos/citología , Enterocitos/metabolismo , Femenino , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Yeyuno/metabolismo , Litostatina/metabolismo , Miembro 2 de la Familia de Transportadores de Soluto 12/metabolismo , Trombospondinas/metabolismo , Proteína Wnt3A/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...