Nogo-A antibody delivery through the olfactory mucosa mitigates experimental autoimmune encephalomyelitis in the mouse CNS.

Pernet, Vincent; Joly, Sandrine; Spiegel, Sebastian; Meli, Ivo; Idriss, Sherif; Maigler, Frank; Mdzomba, Julius Baya; Roenneke, Anna K; Franceschini, Alessandra; Silvestri, Ludovico; Pavone, Francesco S; Calamai, Martino; Schindowski, Katharina; Chan, Andrew (2023). Nogo-A antibody delivery through the olfactory mucosa mitigates experimental autoimmune encephalomyelitis in the mouse CNS. Cell death discovery, 9(1), p. 290. Nature 10.1038/s41420-023-01588-7

[img]
Preview
Text
s41420-023-01588-7.pdf - Published Version
Available under License Creative Commons: Attribution (CC-BY).

Download (4MB) | Preview

Systemic administration of Nogo-A-neutralizing antibody ameliorates experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. However, the blood-brain barrier (BBB) is a major obstacle limiting the passage of systemically applied antibody to the CNS. To bypass the BBB, in the present study we tested the intranasal route of administration by targeting the olfactory mucosa with the Nogo-A-blocking antibody 11C7 mAb in myelin oligodendrocyte glycoprotein-induced EAE. Antibodies were specifically administered onto the olfactory mucosa using a microcatheter. Antibody distribution was examined in the CNS by ELISA and light-sheet microscopy. The effects of 11C7 mAb on Nogo-A signaling were assessed by Western blotting. EAE-induced deficits were monitored daily. Demyelination was observed on spinal cord histological sections. Gene expression changes were followed by trancriptomic analyses. A sensitive capture ELISA revealed a rapid and widespread distribution of 11C7 mAb in the CNS, including the olfactory bulb, the cerebellum and the lumbar spinal cord, but not in the CSF. Light-sheet microscopy allowed to observe antibody accumulation in the parenchyma, thus demonstrating nose-to-brain transfer of IgG. At the functional level, the widespread penetration of 11C7 mAb in the CNS, including the thoracolumbar spinal cord, resulted in the improvement of motor symptoms and in the preservation of myelin in the spinal cord of EAE mice. This was accompanied by Nogo-A signaling downregulation, as reflected by the decreased level of phosphorylated cofilin observed by Western blotting in the cerebellum. In the brain of EAE score-matched animals, 11C7 modified the expression of genes that can influence neurotransmission and cognitive functions, independently of the demyelination phenotype in the spinal cord. In conclusion, our data show the feasibility of olfactory mucosa-directed administration for the delivery of therapeutic antibodies targeting CNS antigens in EAE mice.

Item Type:

Journal Article (Original Article)

Division/Institute:

04 Faculty of Medicine > Department of Head Organs and Neurology (DKNS) > Clinic of Neurology

Graduate School:

Graduate School for Cellular and Biomedical Sciences (GCB)

UniBE Contributor:

Pernet, Vincent, Joly, Sandrine Marina Aline, Spiegel, Sebastian Peter, Meli, Ivo Maurice, Idriss, Sherif Mahmoud Galal, Mdzomba, Julius Baya, Chan, Andrew Hao-Kuang

Subjects:

600 Technology > 610 Medicine & health

ISSN:

2058-7716

Publisher:

Nature

Language:

English

Submitter:

Pubmed Import

Date Deposited:

11 Aug 2023 12:57

Last Modified:

20 Aug 2023 02:37

Publisher DOI:

10.1038/s41420-023-01588-7

PubMed ID:

37558696

BORIS DOI:

10.48350/185354

URI:

https://boris.unibe.ch/id/eprint/185354

Actions (login required)

Edit item Edit item
Provide Feedback