Evaluation of 18F labeled glial fibrillary acidic protein binding nanobody and its brain shuttle peptide fusion proteins using a neuroinflammation rat model

PLoS One. 2023 Jun 14;18(6):e0287047. doi: 10.1371/journal.pone.0287047. eCollection 2023.

Abstract

Astrogliosis is a crucial feature of neuroinflammation and is characterized by the significant upregulation of glial fibrillary acidic protein (GFAP) expression. Hence, visualizing GFAP in the living brain of patients with damaged central nervous system using positron emission tomography (PET) is of great importance, and it is expected to depict neuroinflammation more directly than existing neuroinflammation imaging markers. However, no PET radiotracers for GFAP are currently available. Therefore, neuroimaging with antibody-like affinity proteins could be a viable strategy for visualizing imaging targets that small molecules rarely recognize, such as GFAP, while we need to overcome the challenges of slow clearance and low brain permeability. The E9 nanobody, a small-affinity protein with high affinity and selectivity for GFAP, was utilized in this study. E9 was engineered by fusing a brain shuttle peptide that facilitates blood-brain barrier permeation via two different types of linker domains: E9-GS-ApoE (EGA) and E9-EAK-ApoE (EEA). E9, EGA and EEA were radiolabeled with fluorine-18 using cell-free protein radiosynthesis. In vitro autoradiography showed that all radiolabeled proteins exhibited a significant difference in neuroinflammation in the brain sections created from a rat model constructed by injecting lipopolysaccharide (LPS) into the unilateral striatum of wildtype rats, and an excess competitor displaced their binding. However, exploratory in vivo PET imaging and ex vivo biodistribution studies in the rat model failed to distinguish neuroinflammatory lesions within 3 h of 18F-EEA intravenous injection. This study contributes to a better understanding of the characteristics of small-affinity proteins fused with a brain shuttle peptide for further research into the use of protein molecules as PET tracers for imaging neuropathology.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Apolipoproteins E
  • Brain / diagnostic imaging
  • Glial Fibrillary Acidic Protein
  • Neuroinflammatory Diseases*
  • Peptides
  • Rats
  • Single-Domain Antibodies
  • Tissue Distribution
  • Tomography, X-Ray Computed*

Substances

  • Apolipoproteins E
  • Fluorine-18
  • Glial Fibrillary Acidic Protein
  • Peptides
  • Single-Domain Antibodies

Grants and funding

This study was supported by a grant from Grant-in-Aid for Scientific Research on Innovative Areas (Brain Protein Ageing and Dementia Control) (26117003); Grant-in-Aid for Fostering Joint International Research (B) (19KK0212); Grant-in-Aid for Exploratory Research (20K21564), from the Japan Society for the Promotion of Science and the Ministry of Education, Culture, Sports, Science, and Technology, Japan (MEXT; http://www.mext.go.jp/english/); a Research Grant for Young Scientists from the Division for Interdisciplinary Advanced Research and Education, Tohoku University, Japan (http://www.iiare.tohoku.ac.jp/en/). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.