Cargando…

Epithelial NAD(+) depletion drives mitochondrial dysfunction and contributes to intestinal inflammation

INTRODUCTION: We have previously demonstrated that a pathologic downregulation of peroxisome proliferator-activated receptor–gamma coactivator 1-alpha (PGC1α) within the intestinal epithelium contributes to the pathogenesis of inflammatory bowel disease (IBD). However, the mechanism underlying downr...

Descripción completa

Detalles Bibliográficos
Autores principales: Novak, Elizabeth A., Crawford, Erin C., Mentrup, Heather L., Griffith, Brian D., Fletcher, David M., Flanagan, Meredith R., Schneider, Corinne, Firek, Brian, Rogers, Matthew B., Morowitz, Michael J., Piganelli, Jon D., Wang, Qian, Mollen, Kevin P.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10512956/
https://www.ncbi.nlm.nih.gov/pubmed/37744380
http://dx.doi.org/10.3389/fimmu.2023.1231700
_version_ 1785108471003217920
author Novak, Elizabeth A.
Crawford, Erin C.
Mentrup, Heather L.
Griffith, Brian D.
Fletcher, David M.
Flanagan, Meredith R.
Schneider, Corinne
Firek, Brian
Rogers, Matthew B.
Morowitz, Michael J.
Piganelli, Jon D.
Wang, Qian
Mollen, Kevin P.
author_facet Novak, Elizabeth A.
Crawford, Erin C.
Mentrup, Heather L.
Griffith, Brian D.
Fletcher, David M.
Flanagan, Meredith R.
Schneider, Corinne
Firek, Brian
Rogers, Matthew B.
Morowitz, Michael J.
Piganelli, Jon D.
Wang, Qian
Mollen, Kevin P.
author_sort Novak, Elizabeth A.
collection PubMed
description INTRODUCTION: We have previously demonstrated that a pathologic downregulation of peroxisome proliferator-activated receptor–gamma coactivator 1-alpha (PGC1α) within the intestinal epithelium contributes to the pathogenesis of inflammatory bowel disease (IBD). However, the mechanism underlying downregulation of PGC1α expression and activity during IBD is not yet clear. METHODS: Mice (male; C57Bl/6, Villincre/+;Pgc1afl/fl mice, and Pgc1afl/fl) were subjected to experimental colitis and treated with nicotinamide riboside. Western blot, high-resolution respirometry, nicotinamide adenine dinucleotide (NAD+) quantification, and immunoprecipitation were used to in this study. RESULTS: We demonstrate a significant depletion in the NAD+ levels within the intestinal epithelium of mice undergoing experimental colitis, as well as humans with ulcerative colitis. While we found no decrease in the levels of NAD+-synthesizing enzymes within the intestinal epithelium of mice undergoing experimental colitis, we did find an increase in the mRNA level, as well as the enzymatic activity, of the NAD+-consuming enzyme poly(ADP-ribose) polymerase-1 (PARP1). Treatment of mice undergoing experimental colitis with an NAD+ precursor reduced the severity of colitis, restored mitochondrial function, and increased active PGC1α levels; however, NAD+ repletion did not benefit transgenic mice that lack PGC1α within the intestinal epithelium, suggesting that the therapeutic effects require an intact PGC1α axis. DISCUSSION: Our results emphasize the importance of PGC1α expression to both mitochondrial health and homeostasis within the intestinal epithelium and suggest a novel therapeutic approach for disease management. These findings also provide a mechanistic basis for clinical trials of nicotinamide riboside in IBD patients.
format Online
Article
Text
id pubmed-10512956
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-105129562023-09-22 Epithelial NAD(+) depletion drives mitochondrial dysfunction and contributes to intestinal inflammation Novak, Elizabeth A. Crawford, Erin C. Mentrup, Heather L. Griffith, Brian D. Fletcher, David M. Flanagan, Meredith R. Schneider, Corinne Firek, Brian Rogers, Matthew B. Morowitz, Michael J. Piganelli, Jon D. Wang, Qian Mollen, Kevin P. Front Immunol Immunology INTRODUCTION: We have previously demonstrated that a pathologic downregulation of peroxisome proliferator-activated receptor–gamma coactivator 1-alpha (PGC1α) within the intestinal epithelium contributes to the pathogenesis of inflammatory bowel disease (IBD). However, the mechanism underlying downregulation of PGC1α expression and activity during IBD is not yet clear. METHODS: Mice (male; C57Bl/6, Villincre/+;Pgc1afl/fl mice, and Pgc1afl/fl) were subjected to experimental colitis and treated with nicotinamide riboside. Western blot, high-resolution respirometry, nicotinamide adenine dinucleotide (NAD+) quantification, and immunoprecipitation were used to in this study. RESULTS: We demonstrate a significant depletion in the NAD+ levels within the intestinal epithelium of mice undergoing experimental colitis, as well as humans with ulcerative colitis. While we found no decrease in the levels of NAD+-synthesizing enzymes within the intestinal epithelium of mice undergoing experimental colitis, we did find an increase in the mRNA level, as well as the enzymatic activity, of the NAD+-consuming enzyme poly(ADP-ribose) polymerase-1 (PARP1). Treatment of mice undergoing experimental colitis with an NAD+ precursor reduced the severity of colitis, restored mitochondrial function, and increased active PGC1α levels; however, NAD+ repletion did not benefit transgenic mice that lack PGC1α within the intestinal epithelium, suggesting that the therapeutic effects require an intact PGC1α axis. DISCUSSION: Our results emphasize the importance of PGC1α expression to both mitochondrial health and homeostasis within the intestinal epithelium and suggest a novel therapeutic approach for disease management. These findings also provide a mechanistic basis for clinical trials of nicotinamide riboside in IBD patients. Frontiers Media S.A. 2023-09-07 /pmc/articles/PMC10512956/ /pubmed/37744380 http://dx.doi.org/10.3389/fimmu.2023.1231700 Text en Copyright © 2023 Novak, Crawford, Mentrup, Griffith, Fletcher, Flanagan, Schneider, Firek, Rogers, Morowitz, Piganelli, Wang and Mollen https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
Novak, Elizabeth A.
Crawford, Erin C.
Mentrup, Heather L.
Griffith, Brian D.
Fletcher, David M.
Flanagan, Meredith R.
Schneider, Corinne
Firek, Brian
Rogers, Matthew B.
Morowitz, Michael J.
Piganelli, Jon D.
Wang, Qian
Mollen, Kevin P.
Epithelial NAD(+) depletion drives mitochondrial dysfunction and contributes to intestinal inflammation
title Epithelial NAD(+) depletion drives mitochondrial dysfunction and contributes to intestinal inflammation
title_full Epithelial NAD(+) depletion drives mitochondrial dysfunction and contributes to intestinal inflammation
title_fullStr Epithelial NAD(+) depletion drives mitochondrial dysfunction and contributes to intestinal inflammation
title_full_unstemmed Epithelial NAD(+) depletion drives mitochondrial dysfunction and contributes to intestinal inflammation
title_short Epithelial NAD(+) depletion drives mitochondrial dysfunction and contributes to intestinal inflammation
title_sort epithelial nad(+) depletion drives mitochondrial dysfunction and contributes to intestinal inflammation
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10512956/
https://www.ncbi.nlm.nih.gov/pubmed/37744380
http://dx.doi.org/10.3389/fimmu.2023.1231700
work_keys_str_mv AT novakelizabetha epithelialnaddepletiondrivesmitochondrialdysfunctionandcontributestointestinalinflammation
AT crawforderinc epithelialnaddepletiondrivesmitochondrialdysfunctionandcontributestointestinalinflammation
AT mentrupheatherl epithelialnaddepletiondrivesmitochondrialdysfunctionandcontributestointestinalinflammation
AT griffithbriand epithelialnaddepletiondrivesmitochondrialdysfunctionandcontributestointestinalinflammation
AT fletcherdavidm epithelialnaddepletiondrivesmitochondrialdysfunctionandcontributestointestinalinflammation
AT flanaganmeredithr epithelialnaddepletiondrivesmitochondrialdysfunctionandcontributestointestinalinflammation
AT schneidercorinne epithelialnaddepletiondrivesmitochondrialdysfunctionandcontributestointestinalinflammation
AT firekbrian epithelialnaddepletiondrivesmitochondrialdysfunctionandcontributestointestinalinflammation
AT rogersmatthewb epithelialnaddepletiondrivesmitochondrialdysfunctionandcontributestointestinalinflammation
AT morowitzmichaelj epithelialnaddepletiondrivesmitochondrialdysfunctionandcontributestointestinalinflammation
AT piganellijond epithelialnaddepletiondrivesmitochondrialdysfunctionandcontributestointestinalinflammation
AT wangqian epithelialnaddepletiondrivesmitochondrialdysfunctionandcontributestointestinalinflammation
AT mollenkevinp epithelialnaddepletiondrivesmitochondrialdysfunctionandcontributestointestinalinflammation