Cargando…

Quantification of gamma-secretase modulation differentiates inhibitor compound selectivity between two substrates Notch and amyloid precursor protein

BACKGROUND: Deposition of amyloid-β protein (Aβ) is a major pathological hallmark of Alzheimer's disease (AD). Aβ is generated from γ-secretase cleavage of amyloid precursor protein (APP). In addition to APP, γ-secretase also cleaves other type I integral membrane proteins, including the Notch...

Descripción completa

Detalles Bibliográficos
Autores principales: Yang, Ting, Arslanova, Dilyara, Gu, Yongli, Augelli-Szafran, Corinne, Xia, Weiming
Formato: Texto
Lenguaje:English
Publicado: BioMed Central 2008
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2637266/
https://www.ncbi.nlm.nih.gov/pubmed/18983676
http://dx.doi.org/10.1186/1756-6606-1-15
_version_ 1782164345343442944
author Yang, Ting
Arslanova, Dilyara
Gu, Yongli
Augelli-Szafran, Corinne
Xia, Weiming
author_facet Yang, Ting
Arslanova, Dilyara
Gu, Yongli
Augelli-Szafran, Corinne
Xia, Weiming
author_sort Yang, Ting
collection PubMed
description BACKGROUND: Deposition of amyloid-β protein (Aβ) is a major pathological hallmark of Alzheimer's disease (AD). Aβ is generated from γ-secretase cleavage of amyloid precursor protein (APP). In addition to APP, γ-secretase also cleaves other type I integral membrane proteins, including the Notch receptor, a key molecule involved in embryonic development. RESULTS: To explore selective γ-secretase inhibitors, a combination of five methods was used to systematically determine these inhibitors' profiles on the γ-secretase cleavage of APP and Notch. When two potent γ-secretase inhibitors, compound E (cpd E) and DAPT, were used in a conventional in vitro γ-secretase activity assay, cpd E completely blocked Aβ generation from the cleavage of substrate APP C100, but only had a minor effect on Notch cleavage and NICD generation. Next, cpd E and DAPT were applied to HEK293 cells expressing a truncated Notch substrate NotchΔE. Both cpd E and DAPT were more potent in blocking Aβ generation than NICD generation. Third, a reporter construct was created that carried the NICD targeting promoter with three Su(H) binding sequences followed by the luciferase gene. We found that the inhibition of NICD generation by cpd E and DAPT was consistent with the reduced expression of luciferase gene driven by this Notch targeting promoter. Fourth, levels of "Notch-Aβ-like" (Nβ*) peptide derived from two previously reported chimeric APP with its transmembrane domain or the juxtamembrane portion replaced by the Notch sequence were quantified. Measurement of Nβ* peptides by ELISA confirmed that EC(50)'s of cpd E were much higher for Nβ* than Aβ. Finally, the expression levels of Notch target gene her6 in cpd E or DAPT-treated zebrafish were correlated with the degree of tail curvature due to defective somitogenesis, a well characterized Notch phenotype in zebrafish. CONCLUSION: Our ELISA-based quantification of Aβ and Nβ* in combination with the test in zebrafish provides a novel approach for efficient cell-based screening and in vivo validation of APP selective γ-secretase inhibitors.
format Text
id pubmed-2637266
institution National Center for Biotechnology Information
language English
publishDate 2008
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-26372662009-02-07 Quantification of gamma-secretase modulation differentiates inhibitor compound selectivity between two substrates Notch and amyloid precursor protein Yang, Ting Arslanova, Dilyara Gu, Yongli Augelli-Szafran, Corinne Xia, Weiming Mol Brain Research BACKGROUND: Deposition of amyloid-β protein (Aβ) is a major pathological hallmark of Alzheimer's disease (AD). Aβ is generated from γ-secretase cleavage of amyloid precursor protein (APP). In addition to APP, γ-secretase also cleaves other type I integral membrane proteins, including the Notch receptor, a key molecule involved in embryonic development. RESULTS: To explore selective γ-secretase inhibitors, a combination of five methods was used to systematically determine these inhibitors' profiles on the γ-secretase cleavage of APP and Notch. When two potent γ-secretase inhibitors, compound E (cpd E) and DAPT, were used in a conventional in vitro γ-secretase activity assay, cpd E completely blocked Aβ generation from the cleavage of substrate APP C100, but only had a minor effect on Notch cleavage and NICD generation. Next, cpd E and DAPT were applied to HEK293 cells expressing a truncated Notch substrate NotchΔE. Both cpd E and DAPT were more potent in blocking Aβ generation than NICD generation. Third, a reporter construct was created that carried the NICD targeting promoter with three Su(H) binding sequences followed by the luciferase gene. We found that the inhibition of NICD generation by cpd E and DAPT was consistent with the reduced expression of luciferase gene driven by this Notch targeting promoter. Fourth, levels of "Notch-Aβ-like" (Nβ*) peptide derived from two previously reported chimeric APP with its transmembrane domain or the juxtamembrane portion replaced by the Notch sequence were quantified. Measurement of Nβ* peptides by ELISA confirmed that EC(50)'s of cpd E were much higher for Nβ* than Aβ. Finally, the expression levels of Notch target gene her6 in cpd E or DAPT-treated zebrafish were correlated with the degree of tail curvature due to defective somitogenesis, a well characterized Notch phenotype in zebrafish. CONCLUSION: Our ELISA-based quantification of Aβ and Nβ* in combination with the test in zebrafish provides a novel approach for efficient cell-based screening and in vivo validation of APP selective γ-secretase inhibitors. BioMed Central 2008-11-04 /pmc/articles/PMC2637266/ /pubmed/18983676 http://dx.doi.org/10.1186/1756-6606-1-15 Text en Copyright © 2008 Yang et al; licensee BioMed Central Ltd. http://creativecommons.org/licenses/by/2.0 This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( (http://creativecommons.org/licenses/by/2.0) ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research
Yang, Ting
Arslanova, Dilyara
Gu, Yongli
Augelli-Szafran, Corinne
Xia, Weiming
Quantification of gamma-secretase modulation differentiates inhibitor compound selectivity between two substrates Notch and amyloid precursor protein
title Quantification of gamma-secretase modulation differentiates inhibitor compound selectivity between two substrates Notch and amyloid precursor protein
title_full Quantification of gamma-secretase modulation differentiates inhibitor compound selectivity between two substrates Notch and amyloid precursor protein
title_fullStr Quantification of gamma-secretase modulation differentiates inhibitor compound selectivity between two substrates Notch and amyloid precursor protein
title_full_unstemmed Quantification of gamma-secretase modulation differentiates inhibitor compound selectivity between two substrates Notch and amyloid precursor protein
title_short Quantification of gamma-secretase modulation differentiates inhibitor compound selectivity between two substrates Notch and amyloid precursor protein
title_sort quantification of gamma-secretase modulation differentiates inhibitor compound selectivity between two substrates notch and amyloid precursor protein
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2637266/
https://www.ncbi.nlm.nih.gov/pubmed/18983676
http://dx.doi.org/10.1186/1756-6606-1-15
work_keys_str_mv AT yangting quantificationofgammasecretasemodulationdifferentiatesinhibitorcompoundselectivitybetweentwosubstratesnotchandamyloidprecursorprotein
AT arslanovadilyara quantificationofgammasecretasemodulationdifferentiatesinhibitorcompoundselectivitybetweentwosubstratesnotchandamyloidprecursorprotein
AT guyongli quantificationofgammasecretasemodulationdifferentiatesinhibitorcompoundselectivitybetweentwosubstratesnotchandamyloidprecursorprotein
AT augelliszafrancorinne quantificationofgammasecretasemodulationdifferentiatesinhibitorcompoundselectivitybetweentwosubstratesnotchandamyloidprecursorprotein
AT xiaweiming quantificationofgammasecretasemodulationdifferentiatesinhibitorcompoundselectivitybetweentwosubstratesnotchandamyloidprecursorprotein