Category: Family

Nutrient absorption in the enterocytes

Nutrient absorption in the enterocytes

Diets The Fitness foods are summarised in Supplementary Nutriennt 2. Notably, key genes involved in lipid uptake were not differentially regulated in c-Maf—deficient IECs Fig. Small intestine.

Video

Q\u0026A: Dr. Greger Talks Flax Seeds, Iron Absorption, Mushroom Coffee, and More

Nutrient absorption in the enterocytes -

A minimum of three enteroids in three wells over two independent passages were quantified. Electrophysiological experiments were conducted using a modified Ussing chamber 22 , Mouse jejunum and transplanted HIOs were dissected and immediately placed in ice-cold Krebs-Ringer solution.

Tissues were opened to create a flat epithelial surface. Because seromuscular stripping is associated with release of cyclooxygenases and prostaglandins 22 , and prostaglandins can stimulate L-cells to release GLP1, GLP2 and PYY 37 , we performed the Ussing chamber experiments in intestinal tissue with an intact muscular layer.

Tissues were mounted into sliders 0. D-glucose and Gly-Sar were added to the luminal side of the chamber once the VIP-induced I sc had stabilized at a maximum value.

HIOEs were differentiated for 5—7 days, then were removed from Matrigel and enzymatically dissociated into single-cell suspension using 0. Monolayers were then excised from the plastic Transwell insert and mounted on a glass slide for live confocal Z-stack imaging using a Nikon A1 GaAsP LUNV inverted confocal microscope and NIS Elements software Nikon.

On the final day, enteroids were removed from Matrigel and enzymatically dissociated into single-cell suspension using 0. In all experiments, samples were labeled with either CDH1-mRuby2 or Anti-EpCam-APC BD Biosciences to distinguish epithelial cells and incubated with SYTOX Blue dead cell stain Life Technologies or 7-AAD BD Pharmingen.

Forward scatter and side scatter were used to discriminate doublets and cellular debris. A minimum of 50, events per sample was recorded using an LSR Fortessa flow cytometer BD Biosciences and data were analyzed using FACSDiva software BD Biosciences.

Mice were housed in a specific pathogen-free barrier facility in accordance with NIH Guidelines for the Care and Use of Laboratory Animals. We established a diarrhea score, with 3 representing wet, yellow feces that smeared the perianal fur, and 0 representing normal, dry, brown, well-defined pellets.

Mutant mice which suffered from diarrhea score 3 were included in the rescue experiment. Mice were given access to solid chow on the floor of the cage beginning at postnatal day 10 and weaned at postnatal day Mice were treated daily for a minimum of 10 days after HIOs had been maturing for 8 weeks, then dissected and analyzed.

Data represents measurements taken from individual mice and biological replicates of HIOs from two human pluripotent stem cell lines.

Enteroid experiments were conducted on three independent lines. Further information on research design is available in the Nature Research Reporting Summary linked to this article. All data generated or analyzed during this study are included in the published article and Supplementary Information files.

Source data are available in the Source Data file, and available upon reasonable request from the corresponding author. Gribble, F. Function and mechanisms of enteroendocrine cells and gut hormones in metabolism. Article CAS PubMed Google Scholar.

Wang, J. et al. Mutant neurogenin-3 in congenital malabsorptive diarrhea. Mellitzer, G. Loss of enteroendocrine cells in mice alters lipid absorption and glucose homeostasis and impairs postnatal survival.

Article CAS PubMed PubMed Central Google Scholar. Wright, E. Biology of human sodium glucose transporters. Physiological Rev. Article CAS Google Scholar. Chen, M. Gene ablation for PEPT1 in mice abolishes the effects of dipeptides on small intestinal fluid absorption, short-circuit current, and intracellular pH.

Liver Physiol. Article ADS CAS PubMed Google Scholar. CAS PubMed Google Scholar. Thwaites, D. Gastroenterology , — Burleigh, D. Stimulation of intestinal secretion by vasoactive intestinal peptide and cholera toxin.

Autonomic Neurosci. Yun, C. Natl Acad. Article ADS CAS PubMed PubMed Central Google Scholar. Hyland, N. Functional consequences of neuropeptide Y Y 2 receptor knockout and Y2 antagonism in mouse and human colonic tissues. Cox, H. Peptide YY is critical for acylethanolamine receptor Gprinduced activation of gastrointestinal mucosal responses.

Cell Metab. Tough, I. Endogenous peptide YY and neuropeptide Y inhibit colonic ion transport, contractility and transit differentially via Y1 and Y2 receptors. Moodaley, R. Agonism of free fatty acid receptors 1 and 4 generates peptide YY-mediated inhibitory responses in mouse colon.

Bilchik, A. Peptide YY augments postprandial small intestinal absorption in the conscious dog. Spence, J. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature , —9 Article ADS PubMed Google Scholar. Watson, C. An in vivo model of human small intestine using pluripotent stem cells.

McGrath, P. The basic Helix-Loop-Helix transcription factor NEUROG3 is required for development of the human endocrine pancreas. Diabetes 64 , — Jenny, M. Neurogenin3 is differentially required for endocrine cell fate specification in the intestinal and gastric epithelium.

Embo J. Zhang, X. A comprehensive structure-function study of Neurogenin3 disease-causing alleles during human pancreas and intestinal organoid development. Developmental Cell 50 , — e Dekkers, J. A functional CFTR assay using primary cystic fibrosis intestinal organoids. Foulke-Abel, J.

Human enteroids as a model of upper small intestinal ion transport physiology and pathophysiology. Gastroenterology , — e Article PubMed Google Scholar. Clarke, L. A guide to Ussing chamber studies of mouse intestine.

Anderson, C. Inhibition of intestinal dipeptide transport by the neuropeptide VIP is an anti-absorptive effect via the VPAC1 receptor in a human enterocyte-like cell line Caco Egerod, K. A major lineage of enteroendocrine cells coexpress CCK, secretin, GIP, GLP-1, PYY, and neurotensin but not somatostatin.

Endocrinology , — Bohorquez, D. An enteroendocrine cell-enteric glia connection revealed by 3D electron microscopy. PLoS ONE 9 , e Article ADS PubMed PubMed Central Google Scholar. Neuroepithelial circuit formed by innervation of sensory enteroendocrine cells. Article PubMed PubMed Central Google Scholar.

Mentlein, R. Proteolytic processing of neuropeptide Y and peptide YY by dipeptidyl peptidase IV. Batterham, R. Gut hormone PYY physiologically inhibits food intake. Nature , — McCauley, H. Enteroendocrine regulation of nutrient absorption.

Patel, Y. Somatostatin and its receptor family. Chandran, S. Necrotising enterocolitis in a newborn infant treated with octreotide for chylous effusion: is octreotide safe?

BMJ Case Rep. Endogenous PYY and NPY mediate tonic Y 1 - and Y 2 -mediated absorption in human and mouse colon. Burbank, Los Angeles Cty. Ouchi, R. Modeling steatohepatitis in humans with pluripotent stem cell-derived organoids. Múnera, J. Generation of Gastrointestinal Organoids from Human Pluripotent Stem Cells.

In Organ Regeneration — Springer, Mahe, M. Establishment of human epithelial enteroids and colonoids from whole tissue and biopsy. Matthis, A. Deficient active transport activity in healing mucosa after mild gastric epithelial damage. Briere, D. Activation of Prostaglandin E Receptor 4 Triggers Secretion of Gut Hormone Peptides GLP-1, GLP-2, and PYY.

Endocrinology , 45—53 Moon, C. Development of a primary mouse intestinal epithelial cell monolayer culture system to evaluate factors that modulate IgA transcytosis. Mucosal Immunol. Madisen, L. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain.

Download references. We thank Dr. Gerard Gradwohl and Dr. Mary Estes, Dr. Sarah Blutt and Ms. Xi-Lei Zeng for training in generating HIO-derived enteroid monolayer culture systems; Ms.

Catherine Martini for technical assistance. We acknowledge support provided by the Confocal Imaging Center, the Pluripotent Stem Cell Facility, and Research Flow Cytometry Core at CCHMC. We would like to thank the members of the Wells, Zorn, and Helmrath laboratories for reagents and feedback.

This work was supported by the grants from the NIH, U19 AI J. and the Allen Foundation J. We also received support from the Digestive Disease Research Center P30 DK Heather A.

McCauley, Jacob R. Enriquez, Jonah T. Nichol, J. Guillermo Sanchez, William J. Stone, Michael A. Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Albert Sabin Way, Cincinnati, OH, , USA.

Andrea L. Matthis, Marshall H. You can also search for this author in PubMed Google Scholar. and J. conceived and initiated the project, designed experiments, and wrote the paper, with conceptual input from M.

and E. performed all experiments in collaboration with: J. and W. on mouse transplantation; N. and M. in generating HIO-derived enteroids; A.

on electrophysiological studies. interpreted data. supervised the project. All authors have edited and approved the paper. Correspondence to James M. Peer review information Nature Communications thanks the anonymous reviewer s for their contribution to the peer review of this work.

Open Access This article is licensed under a Creative Commons Attribution 4. Reprints and permissions. Enteroendocrine cells couple nutrient sensing to nutrient absorption by regulating ion transport. Nat Commun 11 , Download citation. Received : 14 May Accepted : 25 August Published : 22 September Anyone you share the following link with will be able to read this content:.

Sorry, a shareable link is not currently available for this article. Provided by the Springer Nature SharedIt content-sharing initiative. By submitting a comment you agree to abide by our Terms and Community Guidelines.

If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate. Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily. Skip to main content Thank you for visiting nature.

nature nature communications articles article. Download PDF. Subjects Gastrointestinal hormones Gastrointestinal models Mechanisms of disease Physiology. Abstract The ability to absorb ingested nutrients is an essential function of all metazoans and utilizes a wide array of nutrient transporters found on the absorptive enterocytes of the small intestine.

Introduction Enteroendocrine cells EECs are a rare population of cells found in the gastrointestinal epithelium that sense nutrients that are passing through the gut and in response secrete more than 20 distinct biologically active peptides. Results The PYY-VIP axis regulates ion transport in small intestine If EECs were required for regulating the normal electrophysiology of the small intestine, we would expect to see deranged ion transport in intestinal tissues lacking EECs.

Full size image. Discussion In this study, we found that loss of all EECs resulted in profound imbalance of ion transport in the small intestine, with subsequent impairment of nutrient absorption. Methods Pluripotent stem cell culture and directed differentiation of HIOs Human embryonic stem cell ESC line WA01 H1 was purchased from WiCell.

In vivo transplant of HIOs 28—35 days after spheroid generation, HIOs were removed from Matrigel and transplanted under the kidney capsule of immune deficient NOD.

Generation and maintenance of HIO-derived enteroids After ~10 weeks of in vivo growth, crypts were isolated from transplanted HIOs and plated in 3D Data availability All data generated or analyzed during this study are included in the published article and Supplementary Information files.

References Gribble, F. Article CAS PubMed Google Scholar Wang, J. Article CAS PubMed Google Scholar Mellitzer, G. Article CAS PubMed PubMed Central Google Scholar Wright, E. Article CAS Google Scholar Chen, M. Article ADS CAS PubMed Google Scholar Wright, E. CAS PubMed Google Scholar Thwaites, D.

Article CAS PubMed Google Scholar Burleigh, D. Article CAS Google Scholar Yun, C. Body weight and temperature were determined using a laboratory scale and an infrared thermometer FTC. All animal experiments were in accordance with the ethical standards of the institution or practice at which the studies were conducted and were reviewed and approved by the responsible ethics committees of Germany LAGeSo.

For the determination of body composition, fat and lean mass were assessed by 1H-magnetic resonance spectroscopy using a Minispec LF50 Body Composition Analyzer Bruker BioSpin.

Basal metabolic parameters were analyzed in a TSE LabMaster System TSE Systems. Mice were acclimated to the metabolic cages individually housed 8 h before starting and supplied with regular diet. Calorimetry was performed with a computer-controlled open circuit calorimetry system composed of 10 metabolic cages.

Each cage was equipped with a special water bottle and a food tray connected to a balance as well as an activity monitor. Parameters were measured for each mouse at 2.

Energy expenditure was adjusted for mouse body weight. Data were analyzed as described Tschӧp et al. IECs were isolated using an adapted protocol from Gracz et al.

Briefly, small intestinal tissue was collected, cut longitudinally, and washed two times in cold PBS before incubating in PBS containing 30 mM EDTA and 1. The tissue was then transferred to PBS containing 30 mM EDTA and incubated under constant stirring for 10 min at 37°C.

RNA was isolated with the RNeasy Micro Kit from Qiagen according to the manufacturer's protocol. RNA libraries were generated and sequencing was performed by Novogene Cambridge, UK.

Three biological replicates of each genotype were sequenced. featureCounts v1. Differential gene expression data were plotted as MA plots using Prism 9 software GraphPad and for selected genes as heatmaps using Morpheus software Broad Institute.

The RNA-Seq data have been deposited to the NCBI GEO platform GSE GSEA was performed using the GSEA tool from the Broad Institute Subramanian et al. Gene sets used in this study were taken from the Kyoto Encyclopedia of Genes and Genomes database or published studies Haber et al.

Serum samples were defrosted, diluted with the extraction solvent, and agitated for 10 min at 1, rpm at room temperature RT; Thermomix Eppendorf. All samples and standards were cooled on ice for 20 min before insoluble matter was removed by centrifugation 2 min, 4°C, 16, rcf.

Amino acids were resolved on a Waters ACQUITY UPLCBEH Amide column 2. Column temperature was 25°C, flow rate 0. Precise source settings and multiple reaction monitoring transitions can be provided upon request. Compounds were identified by matching retention times and fragmentation patterns with analytical pure standards.

Data analysis was performed with Agilent Masshunter software. Signals were integrated and quantified by calibrating with the ratios of natural to isotope-labeled internal standards and adjusted for dilution. Serum amino acid concentrations are reported in micromolars.

IECs were resuspended in µl radioimmunoprecipitation assay RIPA buffer with protease inhibitor and shaken at RT for 15 min Eppendorf thermomixer at rpm. Mouse liver was homogenized in M-Tubes ; Miltenyi Biotec with 5 ml RIPA buffer and protease inhibitor in a GentleMacs instrument.

The protein concentration was determined ; Pierce Protein Assay Kit , and a volume corresponding to 25 µg was transferred to a TwinTec plate Eppendorf , topped up to 50 µl with RIPA before SP3 protein digestion on a Beckmann Biomek i7 workstation as previously described with one-step reduction and alkylation Muller et al.

Briefly, The samples were incubated for 18 min before placing on a magnetic rack for 3 min to pull down the beads with protein.

The reaction was stopped by adding formic acid to a final concentration of 0. Peptide separation was accomplished in a min water to acetonitrile gradient solvent A: 0. The Orbitrap worked in centroid mode with a duty cycle consisting of one MS1 scan at 70, resolution with maximum injection time ms and 3e6 AGC target followed by 40 variable MS2 scans using an 0.

The window length started with 25 MS2 scans at MS source settings were as follows: spray voltage 2. The raw data was processed using DIA-NN 1. MS2 and MS1 mass accuracies were both set to 15 ppm and the scan window size was automatically optimized.

DIA-NN was run in library-free mode with standard settings fasta digest and deep learning-based spectra, retention time and ion mobility prediction using the Uniprot mouse reviewed Swiss-Prot, downloaded on annotations UniProt Consortium, and the match-between-runs option.

Peptide normalized intensities were subjected to quality control with all samples passing acceptance criteria. The missing values of remaining peptides were imputed group-based using the PCA method Josse and Husson, Normalization was performed with LIMMA Ritchie et al.

Statistical analysis of proteomics data was carried out using internally developed R scripts based on publicly available packages. Linear modeling was based on the R package LIMMA Ritchie et al. The categorical factor Class had two levels: Ctrl and Maf ΔIEC IECs.

For finding regulated features, the following criteria were applied: significance level α was set to 0. The log fold-change criterion was applied to guarantee that the measured signal is above the average noise level. Functional GSEA analysis was carried out using R package clusterProfiler Yu et al.

The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE Perez-Riverol et al. Maf ΔIEC and littermate control mice were fasted for 7 h with water ad libitum. Then the body weight of the mice was measured and a total volume in µl of 7.

The amount of glucose in blood was measured before and 30 and 60 min after gavage with a glucometer ACCU-CHEK Mobile. SI tissue was treated with HBSS buffer without calcium and magnesium containing 5 mM EDTA and 10 mM Hepes pH 7.

The supernatant was filtered, and the remaining tissue was mashed through a μm mesh. Recovered cells were counted and stained with different antibodies Table S4.

For flow cytometry, cells were stained with surface antibodies including a viability dye suitable for fixation if required at 4°C for 20 min. Lineage includes anti-CD5, anti-CD8α, anti-CD3, anti-Gr-1, anti-TCRγδ, anti-FcεRIα, anti-CD19, and anti-CD11c. Cells were acquired with a BD LSRFortessa X, and analysis was performed with FlowJo Tree Star software.

To measure the uptake of Cystine by primary epithelial cells, IECs were isolated as described above and incubated with 5 µM BioTracker FITC-Cystine Sigma-Aldrich for 15 min in PBS. The SI was isolated from Maf ΔIEC and littermate control mice.

Afterward, a section of 7 cm of distal small intestine was longitudinally cut and thoroughly washed in cold PBS followed by incubation in PBS containing 5 µM BioTracker FITC-Cystine Sigma-Aldrich for 15 min or 25 µM D-Ala-Lys-AMCA Hycultec for 10 min at 37°C with constant horizontal agitation at rpm.

Finally, nuclei were stained with DAPI for 30 min at RT in combination with FITC-Cystine assay, or with Helix NP Green BioLegend for 15 min at RT in combination with D-Ala-Lys-AMCA assay.

Images were taken on a Zeiss Axio Observer 7 Carl Zeiss and analyzed with ImageJ. For organoid cultures, 20 cm of the proximal SI was collected, cut longitudinally, and washed two times with cold PBS.

The tissue was cut into 2-mm pieces, placed in cold PBS containing 5 mM EDTA, and pipetted up and down 10 times with a disposable pipette coated with 0.

Then supernatant was discarded, and the tissue was incubated twice in 5 mM EDTA in PBS for 10 and 30 min at 4°C with constant agitation.

After EDTA solution removal, fresh PBS was added and the tissue was pipetted up and down 15 times with a disposable pipette coated with 0. This procedure was repeated four more times to obtain a total of five fractions.

The fraction with higher crypt enrichment was identified under the microscope and passed through a µm cell strainer. For Noggin removal experiments, organoids were cultured in ENR medium for 2 d and then ENR was substituted for ER ENR without Noggin.

For crypt expansion index, images of Maf ΔIEC and control organoids were taken for 4 consecutive days after seeding. Buds and total organoids were then counted at least 40 organoids per well using ImageJ software, and crypt expansion index was calculated and shown as the number of crypts per organoid.

mRNA from sorted cells was isolated with the RNeasy Plus Micro Kit according to the manual of the manufacturer QIAGEN. RNA from cell suspensions, organoids, or tissues was extracted using TRIzol reagent following the protocol from ImmGen Heng et al. The isolated RNA was quantified using Nanodrop before qPCR performance.

For IF staining, 5—7 cm of the distal jejunum were taken and Swiss rolls were prepared as previously described with minor adaptations Bialkowska et al. Compound Tissue-Tek, Sakura , and frozen with liquid nitrogen. blocks were cut into 5-µm sections for IF or conventional periodic acid—Schiff staining.

For c-Maf, DCLK1, ChgA, and UEA1 staining, first, slides were rehydrated in cold PBS, then blocked and permeabilized in 0.

Next, slides were incubated with c-Maf antibody in blocking buffer at RT overnight. Then slides were cooled down at RT for 30 min and washed three times in PBS and blocked and permeabilized in blocking buffer at RT for 1 h.

Next, primary antibody staining was performed at RT for 1 h in blocking buffer. Finally, slides were stained with secondary antibodies and DAPI in blocking solution at RT for 1 h and mounted with ProLong Diamond Antifade Mountant Thermo Fisher Scientific.

Images were taken on a confocal microscope LSM Carl Zeiss , a Zeiss Axio Observer 7 Carl Zeiss , and analyzed with ImageJ. For the quantification of SFB, ileal mucosal DNA was isolated.

To specifically isolate the DNA from ileal mucosa, 2 cm of the ileum was taken, cut longitudinally, and washed thoroughly in cold PBS to remove the fecal content.

The clean tissue was washed thoroughly in 0. Bacterial DNA was then isolated from mucosal content with the ZymoBIOMICS DNA Miniprep Kit Zymo Research , and bacterial load was measured by qPCR.

The SFB abundance is presented relative to the abundance of eubacteria. RNA-FISH for SFB was performed as previously described Johansson and Hansson, Images were taken on Zeiss Axio Observer 7 Carl Zeiss and processed using ImageJ. qPCR was performed using a Quant Studio 5 system Applied Biosystems and the SYBR Green PCR Master Mix Kit Applied Biosystems.

The mRNA expression is presented relative to the expression of the housekeeping gene hypoxanthine-guanine phosphoribosyltransferase. Real-time qPCR primer used in this study can be found in Table S3.

A list of antibodies used in this study is provided in Table S4. Data are represented as the means with SEM and summarize or are representative of independent experiments as specified in the text.

Statistical analysis was performed using Prism 9 software GraphPad with two-tailed unpaired Student's t test except RNA-Seq data.

S1 shows data, which demonstrate that intestinal epithelial c-Maf expression is driven by BMP signaling. S2 confirms that the expression of epithelial carbohydrate and protein transporters is reduced in Maf ΔIEC mice.

S3 shows that c-Maf—deficient organoids also express reduced levels of carbohydrate and protein transporters. Table S1 shows DE genes between c-Maf—deficient and control IECs as identified by RNA-Seq.

Table S2 shows differentially expressed proteins between c-Maf—deficient and control IECs and liver tissue as identified by proteomics. Table S3 shows real-time qPCR primers used in this study. Table S4 lists all antibodies used in this study.

We thank Andreas Diefenbach Charité — Universitätsmedizin Berlin, Germany for discussion, providing key resources, and proofreading the manuscript. We further thank Efstathios Stamatiades, Stylianos Gnafakis, Omer Shomrat, Manuela Stäber, Kathrin Textoris-Taube, Roodline Cineus, Ahmed Hegazy, and Frederik Heinrich for resources and technical and experimental help.

The Benjamin Franklin Flow Cytometry Facility and the Core Facility High Throughput Mass Cytometry at Charité — Universitätsmedizin Berlin are greatly acknowledged for cell sorting and proteomics analysis, respectively.

In addition, we thank Dr. Anja A. Kühl and the iPATH facility at Charité — Universitätsmedizin Berlin for support in the preparation of histological samples and staining, the Central Biobank Charité — Universitätsmedizin Berlin for slide scanning and digitalization, and Jörg Piontek for technical support with confocal microscopy.

This research was supported by the Deutsche Forschungsgemeinschaft Priority Program to C. Neumann , by the Ministry of Education and Research as part of the National Research Node "Mass spectrometry in Systems Medicine" under grant agreement L to M.

Author contributions: C. Cosovanu designed and performed most experiments, analyzed data, generated figures, and helped writing the manuscript. Resch helped with experimental design and execution.

Jordan assisted in feeding experiments. Lehmann was responsible for sample preparation for metabolomics and LC-MS instrumentation. Ralser provided funding for personnel and analytical instruments.

Farztdinov performed bioinformatic data analysis and presentation of proteomic data. Mülleder supervised MS measurements and was responsible for MS data analysis and management.

Spranger and S. Brachs supervised metabolic analysis NMR, metabolic cages and provided reagents and equipment for their execution.

Neumann conceived the project, designed and performed experiments, analyzed data, generated figures, and wrote the manuscript. All coauthors read, commented on, and approved the manuscript. shows differentially expressed proteins between c-Maf—deficient and control IECs and liver tissue as identified by proteomics.

c-Maf expression marks mature SI enterocytes of the mid-villus region. A Schematic representation of the murine gastrointestinal tract depicting distinct intestinal segments.

Representative flow cytometric plots of EpCAM vs. c-Maf staining are shown. Numbers in the plots indicate percentage. Scale bar, 50 µm. E Maf expression among distinct SI IEC subsets as determined by scRNA-Seq Haber et al.

F Intensity of c-Maf IF staining along the SI crypt—villus axis. Data are representative of at least two independent experiments. Intestinal epithelial c-Maf expression is driven by BMP signaling.

B SI organoid cultures from Maf ΔIEC and littermate control mice were cultured in ENR medium for 2 d. Afterwards, ENR was refreshed or substituted for ER ENR without Noggin medium.

In addition, organoids were stimulated with BMP-4 for 6 h at day 4 of culture before organoids were harvested for qPCR analysis.

Statistical differences were tested using an unpaired Student's t test two-tailed. Maf ΔIEC mice exhibit a reduced nutritional phenotype. A Representative IF staining of c-Maf and DAPI on cross-section of the SI of Maf ΔIEC and control mice.

B Analysis of c-Maf expression in SI IECs from Maf ΔIEC and control mice. E Regression plot of energy expenditure EE vs. K Representative periodic acid—Schiff staining on cross-section of the SI of Maf ΔIEC and control mice.

L Representative IF staining of DCLK1 and DAPI on cross-section of the SI of Maf ΔIEC and control mice. Data are pooled from at least two independent experiments. Reduced expression of carbohydrate and protein transporters in Maf ΔIEC mice.

B RNA-Seq based PCA of FACS-sorted IECs. Each dot represents an individual biological replicate. H GSEA of whole proteome comparison between liver tissue isolated from Maf ΔIEC and control mice with a focus on biological processes GOBP.

The size of each circle represents the weighted number of proteins involved in the term. NES, normalized enrichment score. Data represent the combined analysis of six and three biologically independent samples from control and Maf ΔIEC mice, respectively.

c-Maf controls intestinal nutrient uptake and sensing. MA plot showing comparison of gene expression between c-Maf—deficient and control IECs. Data represent the combined analysis of three biologically independent samples. C Gene set enrichment plots showing downregulation of gene sets associated with carbohydrate and protein digestion in c-Maf—deficient IECs.

D Representative IF staining of SGLT1 Slc5a1 and PEPT1 Slc15a1 on cross-section of the SI of Maf ΔIEC and control mice. E Volcano plot showing comparison of protein expression between c-Maf—deficient and control IECs.

Data represent the combined analysis of eight and five biologically independent samples from control and Maf ΔIEC mice, respectively. Proteins involved in carbohydrate or protein uptake, whose corresponding genes showed differential expression in our RNA-Seq data, are highlighted.

F GSEA of whole proteome comparison between c-Maf—deficient and control IECs with a focus on biological processes GOBP. H Uptake of FITC-Cystine by primary IECs from Maf ΔIEC and control mice.

Gray peak represents control IECs incubated without FITC-Cystine. Graph on the right shows quantification of FITC-Cystine geometric mean fluorescence intensity gMFI.

I Representative IF microscopy analysis of ex vivo SI whole-tissue uptake assays with fluorescent Biotracker FITC-Cystine or D-Ala-Lys-AMCA.

Scale bar, µm. J Scheme depicting feeding of Maf ΔIEC and control mice with purified diets enriched for carbohydrates C or protein P. F, fat. Reduced expression of carbohydrate and protein transporters in c-Maf—deficient organoids.

C Gating strategy for flow cytometric analysis of SI IEL subsets. Data are representative of least two independent experiments. Epithelial c-Maf deletion affects IELs and SFB colonization.

G Gene set enrichment and heat map plot showing upregulation of genes induced in IECs upon SFB colonization and attachment in c-Maf—deficient IECs Atarashi et al. H qPCR analysis of SFB abundance in ileal mucosal samples from Maf ΔIEC and control mice.

I Representative bacterial FISH staining of SFB on cross-section of the ileum of Maf ΔIEC and control mice. Scale bar, 20 µm. Data are representative of or pooled from at least two independent experiments. c-Maf governs the spatial differentiation and maturation of enterocytes. A Left panel: Visualization of zonation clusters based on the spatial gene expression profiles of enterocytes along the crypt—villus axis Moor et al.

Right panel: Gene set enrichment plots showing up- or downregulation of zonation clusters in c-Maf—deficient IECs.

B Gene set enrichment plots showing up- and downregulation of genes specific for immature and mature enterocytes, respectively, in c-Maf—deficient IECs Haber et al. D Representative microscopy pictures of SI organoids from Maf ΔIEC and littermate control mice at day 4 of culture.

Sign In or Create an Account. Search Dropdown Menu. header search search input Search input auto suggest. filter your search All Content All Journals Journal of Experimental Medicine.

Advanced Search. User Tools Dropdown. Sign In. Skip Nav Destination Article Navigation. Brief Definitive Report September 19 Intestinal epithelial c-Maf expression determines enterocyte differentiation and nutrient uptake in mice In Special Collection: Metabolism Signaling.

Catalina Cosovanu Catalina Cosovanu. This Site. Google Scholar. Philipp Resch Philipp Resch. Stefan Jordan Stefan Jordan. Andrea Lehmann Andrea Lehmann. Markus Ralser Markus Ralser. Vadim Farztdinov Vadim Farztdinov.

Joachim Spranger Joachim Spranger. Michael Mülleder Michael Mülleder. Sebastian Brachs Sebastian Brachs.

Catalina Cosovanu Electrolyte Formula, Philipp Nutrient absorption in the enterocytesStefan JordanAndrea LehmannAlternate-day fasting and mental clarity Ralser enterlcytes, Vadim EntedocytesNuteient SprangerMichael MüllederNutrient absorption in the enterocytes Brachs Natural solutions for water retention, Christian Neumann; Intestinal epithelial c-Maf expression determines enterocyte differentiation and nutrient uptake in mice. J Exp Med 5 December ; 12 : e The primary function of the small intestine SI is to absorb nutrients to maintain whole-body energy homeostasis. Enterocytes are the major epithelial cell type facilitating nutrient sensing and uptake. However, the molecular regulators governing enterocytes have remained undefined. Here, we identify c-Maf as an enterocyte-specific transcription factor within the SI epithelium. Functionally, enterocytes required c-Maf to appropriately differentiate along the villus axis. Thank you for visiting nature. You are using a enterocytex version with limited absorptlon for CSS. Nutrient absorption in the enterocytes obtain the Alternate-day fasting and mental clarity experience, we enterocyfes you Performance tracking through diet a more up to Nutrient absorption in the enterocytes browser or turn off compatibility mode in Internet Explorer. In the meantime, to ensure Nutrisnt support, we are displaying the site without styles and JavaScript. The ability to absorb ingested nutrients is an essential function of all metazoans and utilizes a wide array of nutrient transporters found on the absorptive enterocytes of the small intestine. A unique population of patients has previously been identified with severe congenital malabsorptive diarrhea upon ingestion of any enteral nutrition. The intestines of these patients are macroscopically normal, but lack enteroendocrine cells EECssuggesting an essential role for this rare population of nutrient-sensing cells in regulating macronutrient absorption.

Author: Tygozragore

2 thoughts on “Nutrient absorption in the enterocytes

  1. Nach meiner Meinung lassen Sie den Fehler zu. Geben Sie wir werden es besprechen. Schreiben Sie mir in PM, wir werden reden.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com