SARS-CoV-2 Induces Epithelial-Enteric Neuronal Crosstalk Stimulating VIP Release

Background: Diarrhea is present in up to 30–50% of patients with COVID-19. The mechanism of SARS-CoV-2-induced diarrhea remains unclear. We hypothesized that enterocyte–enteric neuron interactions were important in SARS-CoV-2-induced diarrhea. SARS-CoV-2 induces endoplasmic reticulum (ER) stress in...

Full description

Bibliographic Details
Main Authors: Arun Balasubramaniam, Philip R. Tedbury, Simon M. Mwangi, Yunshan Liu, Ge Li, Didier Merlin, Adam D. Gracz, Peijian He, Stefan G. Sarafianos, Shanthi Srinivasan
Format: Article
Language:English
Published: MDPI AG 2023-01-01
Series:Biomolecules
Subjects:
Online Access:https://www.mdpi.com/2218-273X/13/2/207
_version_ 1797622119487504384
author Arun Balasubramaniam
Philip R. Tedbury
Simon M. Mwangi
Yunshan Liu
Ge Li
Didier Merlin
Adam D. Gracz
Peijian He
Stefan G. Sarafianos
Shanthi Srinivasan
author_facet Arun Balasubramaniam
Philip R. Tedbury
Simon M. Mwangi
Yunshan Liu
Ge Li
Didier Merlin
Adam D. Gracz
Peijian He
Stefan G. Sarafianos
Shanthi Srinivasan
author_sort Arun Balasubramaniam
collection DOAJ
description Background: Diarrhea is present in up to 30–50% of patients with COVID-19. The mechanism of SARS-CoV-2-induced diarrhea remains unclear. We hypothesized that enterocyte–enteric neuron interactions were important in SARS-CoV-2-induced diarrhea. SARS-CoV-2 induces endoplasmic reticulum (ER) stress in enterocytes causing the release of damage associated molecular patterns (DAMPs). The DAMPs then stimulate the release of enteric neurotransmitters that disrupt gut electrolyte homeostasis. Methods: Primary mouse enteric neurons (EN) were exposed to a conditioned medium from ACE2-expressing Caco-2 colonic epithelial cells infected with SARS-CoV-2 or treated with tunicamycin (ER stress inducer). Vasoactive intestinal peptides (VIP) expression and secretion by EN were assessed by RT-PCR and ELISA, respectively. Membrane expression of NHE3 was determined by surface biotinylation. Results: SARS-CoV-2 infection led to increased expression of BiP/GRP78, a marker and key regulator for ER stress in Caco-2 cells. Infected cells secreted the DAMP protein, heat shock protein 70 (HSP70), into the culture media, as revealed by proteomic and Western analyses. The expression of VIP mRNA in EN was up-regulated after treatment with a conditioned medium of SARS-CoV-2-infected Caco-2 cells. CD91, a receptor for HSP70, is abundantly expressed in the cultured mouse EN. Tunicamycin, an inducer of ER stress, also induced the release of HSP70 and Xbp1s, mimicking SARS-CoV-2 infection. Co-treatment of Caco-2 with tunicamycin (apical) and VIP (basolateral) induced a synergistic decrease in membrane expression of Na<sup>+</sup>/H<sup>+</sup> exchanger (NHE3), an important transporter that mediates intestinal Na<sup>+</sup>/fluid absorption. Conclusions: Our findings demonstrate that SARS-CoV-2 enterocyte infection leads to ER stress and the release of DAMPs that up-regulates the expression and release of VIP by EN. VIP in turn inhibits fluid absorption through the downregulation of brush-border membrane expression of NHE3 in enterocytes. These data highlight the role of epithelial-enteric neuronal crosstalk in COVID-19-related diarrhea.
first_indexed 2024-03-11T09:05:48Z
format Article
id doaj.art-40383c3d75514edf99cc8687017e6917
institution Directory Open Access Journal
issn 2218-273X
language English
last_indexed 2024-03-11T09:05:48Z
publishDate 2023-01-01
publisher MDPI AG
record_format Article
series Biomolecules
spelling doaj.art-40383c3d75514edf99cc8687017e69172023-11-16T19:21:52ZengMDPI AGBiomolecules2218-273X2023-01-0113220710.3390/biom13020207SARS-CoV-2 Induces Epithelial-Enteric Neuronal Crosstalk Stimulating VIP ReleaseArun Balasubramaniam0Philip R. Tedbury1Simon M. Mwangi2Yunshan Liu3Ge Li4Didier Merlin5Adam D. Gracz6Peijian He7Stefan G. Sarafianos8Shanthi Srinivasan9Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, GA 30322, USADepartment of Pediatrics, Emory University, Atlanta, GA 30322, USADivision of Digestive Diseases, Department of Medicine, Emory University, Atlanta, GA 30322, USADivision of Digestive Diseases, Department of Medicine, Emory University, Atlanta, GA 30322, USADivision of Digestive Diseases, Department of Medicine, Emory University, Atlanta, GA 30322, USAVA Medical Center Atlanta, Decatur, GA 30033, USADivision of Digestive Diseases, Department of Medicine, Emory University, Atlanta, GA 30322, USADivision of Digestive Diseases, Department of Medicine, Emory University, Atlanta, GA 30322, USADepartment of Pediatrics, Emory University, Atlanta, GA 30322, USADivision of Digestive Diseases, Department of Medicine, Emory University, Atlanta, GA 30322, USABackground: Diarrhea is present in up to 30–50% of patients with COVID-19. The mechanism of SARS-CoV-2-induced diarrhea remains unclear. We hypothesized that enterocyte–enteric neuron interactions were important in SARS-CoV-2-induced diarrhea. SARS-CoV-2 induces endoplasmic reticulum (ER) stress in enterocytes causing the release of damage associated molecular patterns (DAMPs). The DAMPs then stimulate the release of enteric neurotransmitters that disrupt gut electrolyte homeostasis. Methods: Primary mouse enteric neurons (EN) were exposed to a conditioned medium from ACE2-expressing Caco-2 colonic epithelial cells infected with SARS-CoV-2 or treated with tunicamycin (ER stress inducer). Vasoactive intestinal peptides (VIP) expression and secretion by EN were assessed by RT-PCR and ELISA, respectively. Membrane expression of NHE3 was determined by surface biotinylation. Results: SARS-CoV-2 infection led to increased expression of BiP/GRP78, a marker and key regulator for ER stress in Caco-2 cells. Infected cells secreted the DAMP protein, heat shock protein 70 (HSP70), into the culture media, as revealed by proteomic and Western analyses. The expression of VIP mRNA in EN was up-regulated after treatment with a conditioned medium of SARS-CoV-2-infected Caco-2 cells. CD91, a receptor for HSP70, is abundantly expressed in the cultured mouse EN. Tunicamycin, an inducer of ER stress, also induced the release of HSP70 and Xbp1s, mimicking SARS-CoV-2 infection. Co-treatment of Caco-2 with tunicamycin (apical) and VIP (basolateral) induced a synergistic decrease in membrane expression of Na<sup>+</sup>/H<sup>+</sup> exchanger (NHE3), an important transporter that mediates intestinal Na<sup>+</sup>/fluid absorption. Conclusions: Our findings demonstrate that SARS-CoV-2 enterocyte infection leads to ER stress and the release of DAMPs that up-regulates the expression and release of VIP by EN. VIP in turn inhibits fluid absorption through the downregulation of brush-border membrane expression of NHE3 in enterocytes. These data highlight the role of epithelial-enteric neuronal crosstalk in COVID-19-related diarrhea.https://www.mdpi.com/2218-273X/13/2/207SARS-CoV-2diarrheaenterocytesenteric neuronsheat shock protein 70
spellingShingle Arun Balasubramaniam
Philip R. Tedbury
Simon M. Mwangi
Yunshan Liu
Ge Li
Didier Merlin
Adam D. Gracz
Peijian He
Stefan G. Sarafianos
Shanthi Srinivasan
SARS-CoV-2 Induces Epithelial-Enteric Neuronal Crosstalk Stimulating VIP Release
Biomolecules
SARS-CoV-2
diarrhea
enterocytes
enteric neurons
heat shock protein 70
title SARS-CoV-2 Induces Epithelial-Enteric Neuronal Crosstalk Stimulating VIP Release
title_full SARS-CoV-2 Induces Epithelial-Enteric Neuronal Crosstalk Stimulating VIP Release
title_fullStr SARS-CoV-2 Induces Epithelial-Enteric Neuronal Crosstalk Stimulating VIP Release
title_full_unstemmed SARS-CoV-2 Induces Epithelial-Enteric Neuronal Crosstalk Stimulating VIP Release
title_short SARS-CoV-2 Induces Epithelial-Enteric Neuronal Crosstalk Stimulating VIP Release
title_sort sars cov 2 induces epithelial enteric neuronal crosstalk stimulating vip release
topic SARS-CoV-2
diarrhea
enterocytes
enteric neurons
heat shock protein 70
url https://www.mdpi.com/2218-273X/13/2/207
work_keys_str_mv AT arunbalasubramaniam sarscov2inducesepithelialentericneuronalcrosstalkstimulatingviprelease
AT philiprtedbury sarscov2inducesepithelialentericneuronalcrosstalkstimulatingviprelease
AT simonmmwangi sarscov2inducesepithelialentericneuronalcrosstalkstimulatingviprelease
AT yunshanliu sarscov2inducesepithelialentericneuronalcrosstalkstimulatingviprelease
AT geli sarscov2inducesepithelialentericneuronalcrosstalkstimulatingviprelease
AT didiermerlin sarscov2inducesepithelialentericneuronalcrosstalkstimulatingviprelease
AT adamdgracz sarscov2inducesepithelialentericneuronalcrosstalkstimulatingviprelease
AT peijianhe sarscov2inducesepithelialentericneuronalcrosstalkstimulatingviprelease
AT stefangsarafianos sarscov2inducesepithelialentericneuronalcrosstalkstimulatingviprelease
AT shanthisrinivasan sarscov2inducesepithelialentericneuronalcrosstalkstimulatingviprelease