Effective targeting of microglial P2X7 following intracerebroventricular delivery of nanobodies and nanobody-encoding AAVs

Standard

Effective targeting of microglial P2X7 following intracerebroventricular delivery of nanobodies and nanobody-encoding AAVs. / Pinto-Espinoza, Carolina; Guillou, Charlotte; Rissiek, Björn; Wilmes, Maximilian; Javidi, Ehsan; Schwarz, Nicole; Junge, Marten; Haag, Friedrich; Liaukouskaya, Nastassia; Wanner, Nicola; Nicke, Annette; Stortelers, Catelijne; Tan, Yossan-Var; Adriouch, Sahil; Magnus, Tim; Koch-Nolte, Friedrich.

in: FRONT PHARMACOL, Jahrgang 13, 1029236, 2022.

Publikationen: SCORING: Beitrag in Fachzeitschrift/ZeitungSCORING: ZeitschriftenaufsatzForschungBegutachtung

Harvard

APA

Vancouver

Bibtex

@article{f6e8bef942b24af1a871bd91a9cc003d,
title = "Effective targeting of microglial P2X7 following intracerebroventricular delivery of nanobodies and nanobody-encoding AAVs",
abstract = "The P2X7 ion channel is a key sensor for extracellular ATP and a key trigger of sterile inflammation. Intravenous injection of nanobodies that block P2X7 has shown to be beneficial in mouse models of systemic inflammation. P2X7 has also emerged as an attractive therapeutic target for inflammatory brain diseases. However, little is known about the ability of nanobodies to cross the BBB. Here we evaluated the ability of P2X7-specific nanobodies to reach and to block P2X7 on microglia following intravenous or intracerebral administration. For this study, we reformatted and sequence-optimized P2X7 nanobodies for higher stability and elevated isoelectric point. Following injection of nanobodies or nanobody-encoding adeno-associated viral vectors (AAV), we monitored the occupancy and blockade of microglial P2X7 in vivo using ex vivo flow cytometry. Our results show that P2X7 on microglia was within minutes completely occupied and blocked by intracerebroventricularly injected nanobodies, even at low doses. In contrast, very high doses were required to achieve similar effects when injected intravenously. The endogenous production of P2X7-antagonistic nanobodies following intracerebral or intramuscular injection of nanobody-encoding AAVs resulted in a long-term occupancy and blockade of P2X7 on microglia. Our results provide new insights into the conditions for the delivery of nanobodies to microglial P2X7 and point to AAV-mediated delivery of P2X7 nanobodies as a promising strategy for the treatment of sterile brain inflammation.",
author = "Carolina Pinto-Espinoza and Charlotte Guillou and Bj{\"o}rn Rissiek and Maximilian Wilmes and Ehsan Javidi and Nicole Schwarz and Marten Junge and Friedrich Haag and Nastassia Liaukouskaya and Nicola Wanner and Annette Nicke and Catelijne Stortelers and Yossan-Var Tan and Sahil Adriouch and Tim Magnus and Friedrich Koch-Nolte",
note = "Copyright {\textcopyright} 2022 Pinto-Espinoza, Guillou, Rissiek, Wilmes, Javidi, Schwarz, Junge, Haag, Liaukouskaya, Wanner, Nicke, Stortelers, Tan, Adriouch, Magnus and Koch-Nolte.",
year = "2022",
doi = "10.3389/fphar.2022.1029236",
language = "English",
volume = "13",
journal = "FRONT PHARMACOL",
issn = "1663-9812",
publisher = "Frontiers Media S. A.",

}

RIS

TY - JOUR

T1 - Effective targeting of microglial P2X7 following intracerebroventricular delivery of nanobodies and nanobody-encoding AAVs

AU - Pinto-Espinoza, Carolina

AU - Guillou, Charlotte

AU - Rissiek, Björn

AU - Wilmes, Maximilian

AU - Javidi, Ehsan

AU - Schwarz, Nicole

AU - Junge, Marten

AU - Haag, Friedrich

AU - Liaukouskaya, Nastassia

AU - Wanner, Nicola

AU - Nicke, Annette

AU - Stortelers, Catelijne

AU - Tan, Yossan-Var

AU - Adriouch, Sahil

AU - Magnus, Tim

AU - Koch-Nolte, Friedrich

N1 - Copyright © 2022 Pinto-Espinoza, Guillou, Rissiek, Wilmes, Javidi, Schwarz, Junge, Haag, Liaukouskaya, Wanner, Nicke, Stortelers, Tan, Adriouch, Magnus and Koch-Nolte.

PY - 2022

Y1 - 2022

N2 - The P2X7 ion channel is a key sensor for extracellular ATP and a key trigger of sterile inflammation. Intravenous injection of nanobodies that block P2X7 has shown to be beneficial in mouse models of systemic inflammation. P2X7 has also emerged as an attractive therapeutic target for inflammatory brain diseases. However, little is known about the ability of nanobodies to cross the BBB. Here we evaluated the ability of P2X7-specific nanobodies to reach and to block P2X7 on microglia following intravenous or intracerebral administration. For this study, we reformatted and sequence-optimized P2X7 nanobodies for higher stability and elevated isoelectric point. Following injection of nanobodies or nanobody-encoding adeno-associated viral vectors (AAV), we monitored the occupancy and blockade of microglial P2X7 in vivo using ex vivo flow cytometry. Our results show that P2X7 on microglia was within minutes completely occupied and blocked by intracerebroventricularly injected nanobodies, even at low doses. In contrast, very high doses were required to achieve similar effects when injected intravenously. The endogenous production of P2X7-antagonistic nanobodies following intracerebral or intramuscular injection of nanobody-encoding AAVs resulted in a long-term occupancy and blockade of P2X7 on microglia. Our results provide new insights into the conditions for the delivery of nanobodies to microglial P2X7 and point to AAV-mediated delivery of P2X7 nanobodies as a promising strategy for the treatment of sterile brain inflammation.

AB - The P2X7 ion channel is a key sensor for extracellular ATP and a key trigger of sterile inflammation. Intravenous injection of nanobodies that block P2X7 has shown to be beneficial in mouse models of systemic inflammation. P2X7 has also emerged as an attractive therapeutic target for inflammatory brain diseases. However, little is known about the ability of nanobodies to cross the BBB. Here we evaluated the ability of P2X7-specific nanobodies to reach and to block P2X7 on microglia following intravenous or intracerebral administration. For this study, we reformatted and sequence-optimized P2X7 nanobodies for higher stability and elevated isoelectric point. Following injection of nanobodies or nanobody-encoding adeno-associated viral vectors (AAV), we monitored the occupancy and blockade of microglial P2X7 in vivo using ex vivo flow cytometry. Our results show that P2X7 on microglia was within minutes completely occupied and blocked by intracerebroventricularly injected nanobodies, even at low doses. In contrast, very high doses were required to achieve similar effects when injected intravenously. The endogenous production of P2X7-antagonistic nanobodies following intracerebral or intramuscular injection of nanobody-encoding AAVs resulted in a long-term occupancy and blockade of P2X7 on microglia. Our results provide new insights into the conditions for the delivery of nanobodies to microglial P2X7 and point to AAV-mediated delivery of P2X7 nanobodies as a promising strategy for the treatment of sterile brain inflammation.

U2 - 10.3389/fphar.2022.1029236

DO - 10.3389/fphar.2022.1029236

M3 - SCORING: Journal article

C2 - 36299894

VL - 13

JO - FRONT PHARMACOL

JF - FRONT PHARMACOL

SN - 1663-9812

M1 - 1029236

ER -