Kidney organoids reveal redundancy in viral entry pathways during ACE2-dependent SARS-CoV-2 infection

Publikation: Bidrag til tidsskriftTidsskriftartikelForskningfagfællebedømt

Standard

Kidney organoids reveal redundancy in viral entry pathways during ACE2-dependent SARS-CoV-2 infection. / Vanslambrouck, Jessica M.; Neil, Jessica A.; Rudraraju, Rajeev; Mah, Sophia; Tan, Ker Sin; Groenewegen, Ella; Forbes, Thomas A.; Karavendzas, Katerina; Elliott, David A.; Porrello, Enzo R.; Subbarao, Kanta; Little, Melissa H.

I: Journal of Virology, Bind 98, Nr. 3, 2024.

Publikation: Bidrag til tidsskriftTidsskriftartikelForskningfagfællebedømt

Harvard

Vanslambrouck, JM, Neil, JA, Rudraraju, R, Mah, S, Tan, KS, Groenewegen, E, Forbes, TA, Karavendzas, K, Elliott, DA, Porrello, ER, Subbarao, K & Little, MH 2024, 'Kidney organoids reveal redundancy in viral entry pathways during ACE2-dependent SARS-CoV-2 infection', Journal of Virology, bind 98, nr. 3. https://doi.org/10.1128/jvi.01802-23

APA

Vanslambrouck, J. M., Neil, J. A., Rudraraju, R., Mah, S., Tan, K. S., Groenewegen, E., Forbes, T. A., Karavendzas, K., Elliott, D. A., Porrello, E. R., Subbarao, K., & Little, M. H. (2024). Kidney organoids reveal redundancy in viral entry pathways during ACE2-dependent SARS-CoV-2 infection. Journal of Virology, 98(3). https://doi.org/10.1128/jvi.01802-23

Vancouver

Vanslambrouck JM, Neil JA, Rudraraju R, Mah S, Tan KS, Groenewegen E o.a. Kidney organoids reveal redundancy in viral entry pathways during ACE2-dependent SARS-CoV-2 infection. Journal of Virology. 2024;98(3). https://doi.org/10.1128/jvi.01802-23

Author

Vanslambrouck, Jessica M. ; Neil, Jessica A. ; Rudraraju, Rajeev ; Mah, Sophia ; Tan, Ker Sin ; Groenewegen, Ella ; Forbes, Thomas A. ; Karavendzas, Katerina ; Elliott, David A. ; Porrello, Enzo R. ; Subbarao, Kanta ; Little, Melissa H. / Kidney organoids reveal redundancy in viral entry pathways during ACE2-dependent SARS-CoV-2 infection. I: Journal of Virology. 2024 ; Bind 98, Nr. 3.

Bibtex

@article{0513d8734d044947b9c5015dbeff84a7,
title = "Kidney organoids reveal redundancy in viral entry pathways during ACE2-dependent SARS-CoV-2 infection",
abstract = "With a high incidence of acute kidney injury among hospitalized COVID-19 patients, considerable attention has been focussed on whether SARS-CoV-2 specifically targets kidney cells to directly impact renal function, or whether renal damage is primarily an indirect outcome. To date, several studies have utilized kidney organoids to understand the pathogenesis of COVID-19, revealing the ability for SARS-CoV-2 to predominantly infect cells of the proximal tubule (PT), with reduced infectivity following administration of soluble ACE2. However, the immaturity of standard human kidney organoids represents a significant hurdle, leaving the preferred SARS-CoV-2 processing pathway, existence of alternate viral receptors, and the effect of common hypertensive medications on the expression of ACE2 in the context of SARS-CoV-2 exposure incompletely understood. Utilizing a novel kidney organoid model with enhanced PT maturity, genetic- and drug-mediated inhibition of viral entry and processing factors confirmed the requirement for ACE2 for SARS-CoV-2 entry but showed that the virus can utilize dual viral spike protein processing pathways downstream of ACE2 receptor binding. These include TMPRSS- and CTSL/CTSB-mediated non-endosomal and endocytic pathways, with TMPRSS10 likely playing a more significant role in the non-endosomal pathway in renal cells than TMPRSS2. Finally, treatment with the antihypertensive ACE inhibitor, lisinopril, showed negligible impact on receptor expression or susceptibility of renal cells to infection. This study represents the first in-depth characterization of viral entry in stem cell-derived human kidney organoids with enhanced PTs, providing deeper insight into the renal implications of the ongoing COVID-19 pandemic.",
keywords = "COVID-19, kidney, kidney organoids, SARS-CoV-2, stem cells",
author = "Vanslambrouck, {Jessica M.} and Neil, {Jessica A.} and Rajeev Rudraraju and Sophia Mah and Tan, {Ker Sin} and Ella Groenewegen and Forbes, {Thomas A.} and Katerina Karavendzas and Elliott, {David A.} and Porrello, {Enzo R.} and Kanta Subbarao and Little, {Melissa H.}",
note = "Publisher Copyright: {\textcopyright} 2024 Vanslambrouck et al.",
year = "2024",
doi = "10.1128/jvi.01802-23",
language = "English",
volume = "98",
journal = "Journal of Virology",
issn = "0022-538X",
publisher = "American Society for Microbiology",
number = "3",

}

RIS

TY - JOUR

T1 - Kidney organoids reveal redundancy in viral entry pathways during ACE2-dependent SARS-CoV-2 infection

AU - Vanslambrouck, Jessica M.

AU - Neil, Jessica A.

AU - Rudraraju, Rajeev

AU - Mah, Sophia

AU - Tan, Ker Sin

AU - Groenewegen, Ella

AU - Forbes, Thomas A.

AU - Karavendzas, Katerina

AU - Elliott, David A.

AU - Porrello, Enzo R.

AU - Subbarao, Kanta

AU - Little, Melissa H.

N1 - Publisher Copyright: © 2024 Vanslambrouck et al.

PY - 2024

Y1 - 2024

N2 - With a high incidence of acute kidney injury among hospitalized COVID-19 patients, considerable attention has been focussed on whether SARS-CoV-2 specifically targets kidney cells to directly impact renal function, or whether renal damage is primarily an indirect outcome. To date, several studies have utilized kidney organoids to understand the pathogenesis of COVID-19, revealing the ability for SARS-CoV-2 to predominantly infect cells of the proximal tubule (PT), with reduced infectivity following administration of soluble ACE2. However, the immaturity of standard human kidney organoids represents a significant hurdle, leaving the preferred SARS-CoV-2 processing pathway, existence of alternate viral receptors, and the effect of common hypertensive medications on the expression of ACE2 in the context of SARS-CoV-2 exposure incompletely understood. Utilizing a novel kidney organoid model with enhanced PT maturity, genetic- and drug-mediated inhibition of viral entry and processing factors confirmed the requirement for ACE2 for SARS-CoV-2 entry but showed that the virus can utilize dual viral spike protein processing pathways downstream of ACE2 receptor binding. These include TMPRSS- and CTSL/CTSB-mediated non-endosomal and endocytic pathways, with TMPRSS10 likely playing a more significant role in the non-endosomal pathway in renal cells than TMPRSS2. Finally, treatment with the antihypertensive ACE inhibitor, lisinopril, showed negligible impact on receptor expression or susceptibility of renal cells to infection. This study represents the first in-depth characterization of viral entry in stem cell-derived human kidney organoids with enhanced PTs, providing deeper insight into the renal implications of the ongoing COVID-19 pandemic.

AB - With a high incidence of acute kidney injury among hospitalized COVID-19 patients, considerable attention has been focussed on whether SARS-CoV-2 specifically targets kidney cells to directly impact renal function, or whether renal damage is primarily an indirect outcome. To date, several studies have utilized kidney organoids to understand the pathogenesis of COVID-19, revealing the ability for SARS-CoV-2 to predominantly infect cells of the proximal tubule (PT), with reduced infectivity following administration of soluble ACE2. However, the immaturity of standard human kidney organoids represents a significant hurdle, leaving the preferred SARS-CoV-2 processing pathway, existence of alternate viral receptors, and the effect of common hypertensive medications on the expression of ACE2 in the context of SARS-CoV-2 exposure incompletely understood. Utilizing a novel kidney organoid model with enhanced PT maturity, genetic- and drug-mediated inhibition of viral entry and processing factors confirmed the requirement for ACE2 for SARS-CoV-2 entry but showed that the virus can utilize dual viral spike protein processing pathways downstream of ACE2 receptor binding. These include TMPRSS- and CTSL/CTSB-mediated non-endosomal and endocytic pathways, with TMPRSS10 likely playing a more significant role in the non-endosomal pathway in renal cells than TMPRSS2. Finally, treatment with the antihypertensive ACE inhibitor, lisinopril, showed negligible impact on receptor expression or susceptibility of renal cells to infection. This study represents the first in-depth characterization of viral entry in stem cell-derived human kidney organoids with enhanced PTs, providing deeper insight into the renal implications of the ongoing COVID-19 pandemic.

KW - COVID-19

KW - kidney

KW - kidney organoids

KW - SARS-CoV-2

KW - stem cells

U2 - 10.1128/jvi.01802-23

DO - 10.1128/jvi.01802-23

M3 - Journal article

C2 - 38334329

AN - SCOPUS:85188531415

VL - 98

JO - Journal of Virology

JF - Journal of Virology

SN - 0022-538X

IS - 3

ER -

ID: 392661471