Cargando…

Aquaporin 9 induction in human iPSC‐derived hepatocytes facilitates modeling of ornithine transcarbamylase deficiency

BACKGROUND AND AIMS: Patient‐derived human‐induced pluripotent stem cells (hiPSCs) differentiated into hepatocytes (hiPSC‐Heps) have facilitated the study of rare genetic liver diseases. Here, we aimed to establish an in vitro liver disease model of the urea cycle disorder ornithine transcarbamylase...

Descripción completa

Detalles Bibliográficos
Autores principales: Laemmle, Alexander, Poms, Martin, Hsu, Bernadette, Borsuk, Mariia, Rüfenacht, Véronique, Robinson, Joshua, Sadowski, Martin C., Nuoffer, Jean‐Marc, Häberle, Johannes, Willenbring, Holger
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9295321/
https://www.ncbi.nlm.nih.gov/pubmed/34786702
http://dx.doi.org/10.1002/hep.32247
_version_ 1784750036247117824
author Laemmle, Alexander
Poms, Martin
Hsu, Bernadette
Borsuk, Mariia
Rüfenacht, Véronique
Robinson, Joshua
Sadowski, Martin C.
Nuoffer, Jean‐Marc
Häberle, Johannes
Willenbring, Holger
author_facet Laemmle, Alexander
Poms, Martin
Hsu, Bernadette
Borsuk, Mariia
Rüfenacht, Véronique
Robinson, Joshua
Sadowski, Martin C.
Nuoffer, Jean‐Marc
Häberle, Johannes
Willenbring, Holger
author_sort Laemmle, Alexander
collection PubMed
description BACKGROUND AND AIMS: Patient‐derived human‐induced pluripotent stem cells (hiPSCs) differentiated into hepatocytes (hiPSC‐Heps) have facilitated the study of rare genetic liver diseases. Here, we aimed to establish an in vitro liver disease model of the urea cycle disorder ornithine transcarbamylase deficiency (OTCD) using patient‐derived hiPSC‐Heps. APPROACH AND RESULTS: Before modeling OTCD, we addressed the question of why hiPSC‐Heps generally secrete less urea than adult primary human hepatocytes (PHHs). Because hiPSC‐Heps are not completely differentiated and maintain some characteristics of fetal PHHs, we compared gene‐expression levels in human fetal and adult liver tissue to identify genes responsible for reduced urea secretion in hiPSC‐Heps. We found lack of aquaporin 9 (AQP9) expression in fetal liver tissue as well as in hiPSC‐Heps, and showed that forced expression of AQP9 in hiPSC‐Heps restores urea secretion and normalizes the response to ammonia challenge by increasing ureagenesis. Furthermore, we proved functional ureagenesis by challenging AQP9‐expressing hiPSC‐Heps with ammonium chloride labeled with the stable isotope [(15)N] ((15)NH(4)Cl) and by assessing enrichment of [(15)N]‐labeled urea. Finally, using hiPSC‐Heps derived from patients with OTCD, we generated a liver disease model that recapitulates the hepatic manifestation of the human disease. Restoring OTC expression—together with AQP9—was effective in fully correcting OTC activity and normalizing ureagenesis as assessed by (15)NH(4)Cl stable‐isotope challenge. CONCLUSION: Our results identify a critical role for AQP9 in functional urea metabolism and establish the feasibility of in vitro modeling of OTCD with hiPSC‐Heps. By facilitating studies of OTCD genotype/phenotype correlation and drug screens, our model has potential for improving the therapy of OTCD.
format Online
Article
Text
id pubmed-9295321
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-92953212022-10-14 Aquaporin 9 induction in human iPSC‐derived hepatocytes facilitates modeling of ornithine transcarbamylase deficiency Laemmle, Alexander Poms, Martin Hsu, Bernadette Borsuk, Mariia Rüfenacht, Véronique Robinson, Joshua Sadowski, Martin C. Nuoffer, Jean‐Marc Häberle, Johannes Willenbring, Holger Hepatology Original Articles BACKGROUND AND AIMS: Patient‐derived human‐induced pluripotent stem cells (hiPSCs) differentiated into hepatocytes (hiPSC‐Heps) have facilitated the study of rare genetic liver diseases. Here, we aimed to establish an in vitro liver disease model of the urea cycle disorder ornithine transcarbamylase deficiency (OTCD) using patient‐derived hiPSC‐Heps. APPROACH AND RESULTS: Before modeling OTCD, we addressed the question of why hiPSC‐Heps generally secrete less urea than adult primary human hepatocytes (PHHs). Because hiPSC‐Heps are not completely differentiated and maintain some characteristics of fetal PHHs, we compared gene‐expression levels in human fetal and adult liver tissue to identify genes responsible for reduced urea secretion in hiPSC‐Heps. We found lack of aquaporin 9 (AQP9) expression in fetal liver tissue as well as in hiPSC‐Heps, and showed that forced expression of AQP9 in hiPSC‐Heps restores urea secretion and normalizes the response to ammonia challenge by increasing ureagenesis. Furthermore, we proved functional ureagenesis by challenging AQP9‐expressing hiPSC‐Heps with ammonium chloride labeled with the stable isotope [(15)N] ((15)NH(4)Cl) and by assessing enrichment of [(15)N]‐labeled urea. Finally, using hiPSC‐Heps derived from patients with OTCD, we generated a liver disease model that recapitulates the hepatic manifestation of the human disease. Restoring OTC expression—together with AQP9—was effective in fully correcting OTC activity and normalizing ureagenesis as assessed by (15)NH(4)Cl stable‐isotope challenge. CONCLUSION: Our results identify a critical role for AQP9 in functional urea metabolism and establish the feasibility of in vitro modeling of OTCD with hiPSC‐Heps. By facilitating studies of OTCD genotype/phenotype correlation and drug screens, our model has potential for improving the therapy of OTCD. John Wiley and Sons Inc. 2021-12-18 2022-09 /pmc/articles/PMC9295321/ /pubmed/34786702 http://dx.doi.org/10.1002/hep.32247 Text en © 2021 The Authors. Hepatology published by Wiley Periodicals LLC on behalf of American Association for the Study of Liver Diseases https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc-nd/4.0/ (https://creativecommons.org/licenses/by-nc-nd/4.0/) License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.
spellingShingle Original Articles
Laemmle, Alexander
Poms, Martin
Hsu, Bernadette
Borsuk, Mariia
Rüfenacht, Véronique
Robinson, Joshua
Sadowski, Martin C.
Nuoffer, Jean‐Marc
Häberle, Johannes
Willenbring, Holger
Aquaporin 9 induction in human iPSC‐derived hepatocytes facilitates modeling of ornithine transcarbamylase deficiency
title Aquaporin 9 induction in human iPSC‐derived hepatocytes facilitates modeling of ornithine transcarbamylase deficiency
title_full Aquaporin 9 induction in human iPSC‐derived hepatocytes facilitates modeling of ornithine transcarbamylase deficiency
title_fullStr Aquaporin 9 induction in human iPSC‐derived hepatocytes facilitates modeling of ornithine transcarbamylase deficiency
title_full_unstemmed Aquaporin 9 induction in human iPSC‐derived hepatocytes facilitates modeling of ornithine transcarbamylase deficiency
title_short Aquaporin 9 induction in human iPSC‐derived hepatocytes facilitates modeling of ornithine transcarbamylase deficiency
title_sort aquaporin 9 induction in human ipsc‐derived hepatocytes facilitates modeling of ornithine transcarbamylase deficiency
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9295321/
https://www.ncbi.nlm.nih.gov/pubmed/34786702
http://dx.doi.org/10.1002/hep.32247
work_keys_str_mv AT laemmlealexander aquaporin9inductioninhumanipscderivedhepatocytesfacilitatesmodelingofornithinetranscarbamylasedeficiency
AT pomsmartin aquaporin9inductioninhumanipscderivedhepatocytesfacilitatesmodelingofornithinetranscarbamylasedeficiency
AT hsubernadette aquaporin9inductioninhumanipscderivedhepatocytesfacilitatesmodelingofornithinetranscarbamylasedeficiency
AT borsukmariia aquaporin9inductioninhumanipscderivedhepatocytesfacilitatesmodelingofornithinetranscarbamylasedeficiency
AT rufenachtveronique aquaporin9inductioninhumanipscderivedhepatocytesfacilitatesmodelingofornithinetranscarbamylasedeficiency
AT robinsonjoshua aquaporin9inductioninhumanipscderivedhepatocytesfacilitatesmodelingofornithinetranscarbamylasedeficiency
AT sadowskimartinc aquaporin9inductioninhumanipscderivedhepatocytesfacilitatesmodelingofornithinetranscarbamylasedeficiency
AT nuofferjeanmarc aquaporin9inductioninhumanipscderivedhepatocytesfacilitatesmodelingofornithinetranscarbamylasedeficiency
AT haberlejohannes aquaporin9inductioninhumanipscderivedhepatocytesfacilitatesmodelingofornithinetranscarbamylasedeficiency
AT willenbringholger aquaporin9inductioninhumanipscderivedhepatocytesfacilitatesmodelingofornithinetranscarbamylasedeficiency