Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros

Banco de datos
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Front Pharmacol ; 13: 1029236, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36299894

RESUMEN

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.

2.
Front Immunol ; 12: 704408, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34489954

RESUMEN

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.


Asunto(s)
ADP Ribosa Transferasas , Productos Biológicos/inmunología , Dependovirus , Vectores Genéticos , Depleción Linfocítica , Receptores Purinérgicos P2X7/inmunología , Anticuerpos de Dominio Único , ADP Ribosa Transferasas/antagonistas & inhibidores , ADP Ribosa Transferasas/inmunología , Animales , Ratones , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología
3.
Methods Mol Biol ; 2041: 117-136, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31646484

RESUMEN

Mammalian purinoreceptors respond to extracellular nucleotides and their metabolites, for example, following the release of ATP or NAD+ from cells and their hydrolysis by ectonucleotidases. Membrane purinoreceptors are expressed as ionotropic ligand-gated ion channels designated P2X receptors, or as metabotropic G-protein coupled receptors designated P1 or P2Y receptors, on the cell surface of different cell types. In this chapter, we provide protocols to monitor the expression and activity of purinoreceptors on the cell membrane of living cells by flow cytometry.


Asunto(s)
Adenosina Trifosfato/metabolismo , Membrana Celular/metabolismo , Citometría de Flujo/métodos , Nucleótidos/metabolismo , Receptores Purinérgicos/metabolismo , Animales , Humanos , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA