Your browser doesn't support javascript.
loading
Evaluation of 18F labeled glial fibrillary acidic protein binding nanobody and its brain shuttle peptide fusion proteins using a neuroinflammation rat model.
Morito, Takahiro; Harada, Ryuichi; Iwata, Ren; Ishikawa, Yoichi; Okamura, Nobuyuki; Kudo, Yukitsuka; Furumoto, Shozo; Yanai, Kazuhiko; Tashiro, Manabu.
Afiliación
  • Morito T; Division of Cyclotron Nuclear Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.
  • Harada R; Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.
  • Iwata R; Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.
  • Ishikawa Y; Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan.
  • Okamura N; Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai, Miyagi, Japan.
  • Kudo Y; Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai, Miyagi, Japan.
  • Furumoto S; Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan.
  • Yanai K; Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan.
  • Tashiro M; Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai, Miyagi, Japan.
PLoS One ; 18(6): e0287047, 2023.
Article en En | MEDLINE | ID: mdl-37315033
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.
Asunto(s)

Texto completo: 1 Base de datos: MEDLINE Asunto principal: Tomografía Computarizada por Rayos X / Enfermedades Neuroinflamatorias Tipo de estudio: Prognostic_studies Idioma: En Revista: PLoS One Asunto de la revista: CIENCIA / MEDICINA Año: 2023 Tipo del documento: Article

Texto completo: 1 Base de datos: MEDLINE Asunto principal: Tomografía Computarizada por Rayos X / Enfermedades Neuroinflamatorias Tipo de estudio: Prognostic_studies Idioma: En Revista: PLoS One Asunto de la revista: CIENCIA / MEDICINA Año: 2023 Tipo del documento: Article