A Methodological Approach Using rAAV Vectors Encoding Nanobody-Based Biologics to Evaluate ARTC2.2 and P2X7 In Vivo

Standard

A Methodological Approach Using rAAV Vectors Encoding Nanobody-Based Biologics to Evaluate ARTC2.2 and P2X7 In Vivo. / Gondé, Henri; Demeules, Mélanie; Hardet, Romain; Scarpitta, Allan; Junge, Marten; Pinto-Espinoza, Carolina; Varin, Rémi; Koch-Nolte, Friedrich; Boyer, Olivier; Adriouch, Sahil.

In: FRONT IMMUNOL, Vol. 12, 704408, 2021.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

Gondé, H, Demeules, M, Hardet, R, Scarpitta, A, Junge, M, Pinto-Espinoza, C, Varin, R, Koch-Nolte, F, Boyer, O & Adriouch, S 2021, 'A Methodological Approach Using rAAV Vectors Encoding Nanobody-Based Biologics to Evaluate ARTC2.2 and P2X7 In Vivo', FRONT IMMUNOL, vol. 12, 704408. https://doi.org/10.3389/fimmu.2021.704408

APA

Gondé, H., Demeules, M., Hardet, R., Scarpitta, A., Junge, M., Pinto-Espinoza, C., Varin, R., Koch-Nolte, F., Boyer, O., & Adriouch, S. (2021). A Methodological Approach Using rAAV Vectors Encoding Nanobody-Based Biologics to Evaluate ARTC2.2 and P2X7 In Vivo. FRONT IMMUNOL, 12, [704408]. https://doi.org/10.3389/fimmu.2021.704408

Vancouver

Bibtex

@article{1d856735b792497585686854b41d434f,
title = "A Methodological Approach Using rAAV Vectors Encoding Nanobody-Based Biologics to Evaluate ARTC2.2 and P2X7 In Vivo",
abstract = "On murine T cells, mono-ADP ribosyltransferase ARTC2.2 catalyzes ADP-ribosylation of various surface proteins when nicotinamide adenine dinucleotide (NAD+) is released into the extracellular compartment. Covalent ADP-ribosylation of the P2X7 receptor by ARTC2.2 thereby represents an additional mechanism of activation, complementary to its triggering by extracellular ATP. P2X7 is a multifaceted receptor that may represents a potential target in inflammatory, and neurodegenerative diseases, as well as in cancer. We present herein an experimental approach using intramuscular injection of recombinant AAV vectors (rAAV) encoding nanobody-based biologics targeting ARTC2.2 or P2X7. We demonstrate the ability of these in vivo generated biologics to potently and durably block P2X7 or ARTC2.2 activities in vivo, or in contrast, to potentiate NAD+- or ATP-induced activation of P2X7. We additionally demonstrate the ability of rAAV-encoded functional heavy chain antibodies to elicit long-term depletion of T cells expressing high levels of ARTC2.2 or P2X7. Our approach of using rAAV to generate functional nanobody-based biologics in vivo appears promising to evaluate the role of ARTC2.2 and P2X7 in murine acute as well as chronic disease models.",
keywords = "ADP Ribose Transferases/antagonists & inhibitors, Animals, Biological Products/immunology, Dependovirus, Genetic Vectors, Lymphocyte Depletion, Mice, Receptors, Purinergic P2X7/immunology, Single-Domain Antibodies/genetics",
author = "Henri Gond{\'e} and M{\'e}lanie Demeules and Romain Hardet and Allan Scarpitta and Marten Junge and Carolina Pinto-Espinoza and R{\'e}mi Varin and Friedrich Koch-Nolte and Olivier Boyer and Sahil Adriouch",
note = "Copyright {\textcopyright} 2021 Gond{\'e}, Demeules, Hardet, Scarpitta, Junge, Pinto-Espinoza, Varin, Koch-Nolte, Boyer and Adriouch.",
year = "2021",
doi = "10.3389/fimmu.2021.704408",
language = "English",
volume = "12",
journal = "FRONT IMMUNOL",
issn = "1664-3224",
publisher = "Lausanne : Frontiers Research Foundation",

}

RIS

TY - JOUR

T1 - A Methodological Approach Using rAAV Vectors Encoding Nanobody-Based Biologics to Evaluate ARTC2.2 and P2X7 In Vivo

AU - Gondé, Henri

AU - Demeules, Mélanie

AU - Hardet, Romain

AU - Scarpitta, Allan

AU - Junge, Marten

AU - Pinto-Espinoza, Carolina

AU - Varin, Rémi

AU - Koch-Nolte, Friedrich

AU - Boyer, Olivier

AU - Adriouch, Sahil

N1 - Copyright © 2021 Gondé, Demeules, Hardet, Scarpitta, Junge, Pinto-Espinoza, Varin, Koch-Nolte, Boyer and Adriouch.

PY - 2021

Y1 - 2021

N2 - On murine T cells, mono-ADP ribosyltransferase ARTC2.2 catalyzes ADP-ribosylation of various surface proteins when nicotinamide adenine dinucleotide (NAD+) is released into the extracellular compartment. Covalent ADP-ribosylation of the P2X7 receptor by ARTC2.2 thereby represents an additional mechanism of activation, complementary to its triggering by extracellular ATP. P2X7 is a multifaceted receptor that may represents a potential target in inflammatory, and neurodegenerative diseases, as well as in cancer. We present herein an experimental approach using intramuscular injection of recombinant AAV vectors (rAAV) encoding nanobody-based biologics targeting ARTC2.2 or P2X7. We demonstrate the ability of these in vivo generated biologics to potently and durably block P2X7 or ARTC2.2 activities in vivo, or in contrast, to potentiate NAD+- or ATP-induced activation of P2X7. We additionally demonstrate the ability of rAAV-encoded functional heavy chain antibodies to elicit long-term depletion of T cells expressing high levels of ARTC2.2 or P2X7. Our approach of using rAAV to generate functional nanobody-based biologics in vivo appears promising to evaluate the role of ARTC2.2 and P2X7 in murine acute as well as chronic disease models.

AB - On murine T cells, mono-ADP ribosyltransferase ARTC2.2 catalyzes ADP-ribosylation of various surface proteins when nicotinamide adenine dinucleotide (NAD+) is released into the extracellular compartment. Covalent ADP-ribosylation of the P2X7 receptor by ARTC2.2 thereby represents an additional mechanism of activation, complementary to its triggering by extracellular ATP. P2X7 is a multifaceted receptor that may represents a potential target in inflammatory, and neurodegenerative diseases, as well as in cancer. We present herein an experimental approach using intramuscular injection of recombinant AAV vectors (rAAV) encoding nanobody-based biologics targeting ARTC2.2 or P2X7. We demonstrate the ability of these in vivo generated biologics to potently and durably block P2X7 or ARTC2.2 activities in vivo, or in contrast, to potentiate NAD+- or ATP-induced activation of P2X7. We additionally demonstrate the ability of rAAV-encoded functional heavy chain antibodies to elicit long-term depletion of T cells expressing high levels of ARTC2.2 or P2X7. Our approach of using rAAV to generate functional nanobody-based biologics in vivo appears promising to evaluate the role of ARTC2.2 and P2X7 in murine acute as well as chronic disease models.

KW - ADP Ribose Transferases/antagonists & inhibitors

KW - Animals

KW - Biological Products/immunology

KW - Dependovirus

KW - Genetic Vectors

KW - Lymphocyte Depletion

KW - Mice

KW - Receptors, Purinergic P2X7/immunology

KW - Single-Domain Antibodies/genetics

U2 - 10.3389/fimmu.2021.704408

DO - 10.3389/fimmu.2021.704408

M3 - SCORING: Journal article

C2 - 34489954

VL - 12

JO - FRONT IMMUNOL

JF - FRONT IMMUNOL

SN - 1664-3224

M1 - 704408

ER -