Original Research
Human Intestinal Enteroids With Inducible Neurogenin-3 Expression as a Novel Model of Gut Hormone Secretion

https://doi.org/10.1016/j.jcmgh.2019.04.010Get rights and content
Under a Creative Commons license
open access

Background & Aims

Enteroendocrine cells (EECs) are specialized epithelial cells that produce molecules vital for intestinal homeostasis, but because of their limited numbers, in-depth functional studies have remained challenging. Human intestinal enteroids (HIEs) that are derived from intestinal crypt stem cells are biologically relevant in an in vitro model of the intestinal epithelium. HIEs contain all intestinal epithelial cell types; however, similar to the intestine, HIEs spontaneously produce few EECs, which limits their study.

Methods

To increase the number of EECs in HIEs, we used lentivirus transduction to stably engineer jejunal HIEs with doxycycline-inducible expression of neurogenin-3 (NGN3), a transcription factor that drives EEC differentiation (tetNGN3-HIEs). We examined the impact of NGN3 induction on EECs by quantifying the increase in the enterochromaffin cells and other EEC subtypes. We functionally assessed secretion of serotonin and EEC hormones in response to norepinephrine and rotavirus infection.

Results

Treating tetNGN3-HIEs with doxycycline induced a dose-dependent increase of chromogranin A (ChgA)-positive and serotonin-positive cells, showing increased enterochromaffin cell differentiation. Despite increased ChgA-positive cells, other differentiated cell types of the epithelium remained largely unchanged by gene expression and immunostaining. RNA sequencing of doxycycline-induced tetNGN3-HIEs identified increased expression of key hormones and enzymes associated with several other EEC subtypes. Doxycycline-induced tetNGN3-HIEs secreted serotonin, monocyte chemoattractant protein-1, glucose-dependent insulinotropic peptide, peptide YY, and ghrelin in response to norepinephrine and rotavirus infection, further supporting the presence of multiple EEC types.

Conclusions

We have combined HIEs and inducible-NGN3 expression to establish a flexible in vitro model system for functional studies of EECs in enteroids and advance the molecular and physiological investigation of EECs.

Keywords

Enteroendocrine Cell
Enteroid
Serotonin

Abbreviations used in this paper

cDNA
complementary DNA
ChgA
chromogranin A
CMGF-
complete medium without growth factors
CMGF+
complete media with growth factors
DAPI
4′,6-diamidino-2-phenylindole
DIC
differential interference contrast
DMEM
Dulbecco’s modified Eagle medium
EEC
enteroendocrine cell
GI
gastrointestinal
GIP
glucose-dependent insulinotropic peptide
GLP-1
glucagon-like peptide-1
HIE
human intestinal enteroid
HIO
human intestinal organoid
hpi
hours postinfection
hW-CMGF+
high Wnt complete media with growth factors
IL
interleukin
MCP-1
monocyte chemoattractant protein-1
mRNA
messenger RNA
NGN3
neurogenin-3
PBS
phosphate-buffered saline
PP
pancreatic polypeptide
PYY
peptide YY
qPCR
quantitative polymerase chain reaction
RV
rotavirus
SI
sucrase isomaltase
tet
tetracycline
3D
3-dimensional
TNF-α
tumor necrosis factor-α
2D
2-dimensional
VIL1
villin

Cited by (0)

Author contributions Alexandra L. Chang-Graham, Heather A. Danhof, Melinda A. Engevik, Catherine Tomaro-Duchesneau, Umesh C. Karandikar, Mary K. Estes, Robert A. Britton, and Joseph M. Hyser were responsible for the concept and design; Alexandra L. Chang-Graham, Heather A. Danhof, Melinda A. Engevik, Catherine Tomaro-Duchesneau, Umesh C. Karandikar, Mary K. Estes, James Versalovic, Robert A. Britton, and Joseph M. Hyser provided intellectual contributions; Alexandra L. Chang-Graham, Heather A. Danhof, Melinda A. Engevik, Catherine Tomaro-Duchesneau, and Umesh C. Karandikar acquired data; Alexandra L. Chang-Graham, Heather A. Danhof, Melinda A. Engevik, Catherine Tomaro-Duchesneau, and Umesh C. Karandikar performed the data analysis, statistical analysis, and interpretation; Alexandra L. Chang-Graham, Heather A. Danhof, Melinda A. Engevik, and Joseph M. Hyser drafted and edited the manuscript; and Mary K. Estes, James Versalovic, Robert A. Britton, and Joseph M. Hyser obtained funding.

Conflicts of interest The authors disclose no conflicts.

Funding Supported by National Institutes of Health grants F30 DK112563 (A.C.G), U01 CA170930 (J.V.), R01 DK103759 (R.A.B), U19 AI116497, R01 AI080656 (M.K.E.), R03 DK110270, and R01 DK115507 (J.M.H.); Baylor College of Medicine Seed Funding (J.M.H.); and Fonds de Recherche Santé Québec (C.T.D.). This project also was supported in part by the National Institutes of Health Public Health Service (PHS) grant P30 DK056338 for the Texas Medical Center Digestive Diseases Center. Additional funding support for the Baylor College of Medicine Integrated Microscopy Core was provided by the National Institutes of Health (DK56338, CA125123), Cancer Prevention and Research Institute of Texas (RP150578, RP170719), the Dan L. Duncan Comprehensive Cancer Center, and the John S. Dunn Gulf Coast Consortium for Chemical Genomics.

Authors share co-first authorship.