Please use this identifier to cite or link to this item: https://doi.org/10.1186/1742-2094-9-62
DC FieldValue
dc.titleMinocycline corrects early, pre-plaque neuroinflammation and inhibits BACE-1 in a transgenic model of Alzheimer's disease-like amyloid pathology
dc.contributor.authorFerretti, M.T
dc.contributor.authorAllard, S
dc.contributor.authorPartridge, V
dc.contributor.authorDucatenzeiler, A
dc.contributor.authorCuello, A.C
dc.date.accessioned2020-10-20T08:06:56Z
dc.date.available2020-10-20T08:06:56Z
dc.date.issued2012
dc.identifier.citationFerretti, M.T, Allard, S, Partridge, V, Ducatenzeiler, A, Cuello, A.C (2012). Minocycline corrects early, pre-plaque neuroinflammation and inhibits BACE-1 in a transgenic model of Alzheimer's disease-like amyloid pathology. Journal of Neuroinflammation 9 : 62. ScholarBank@NUS Repository. https://doi.org/10.1186/1742-2094-9-62
dc.identifier.issn1742-2094
dc.identifier.urihttps://scholarbank.nus.edu.sg/handle/10635/178149
dc.description.abstractBackground: A growing body of evidence indicates that inflammation is one of the earliest neuropathological events in Alzheimer's disease. Accordingly, we have recently shown the occurrence of an early, pro-inflammatory reaction in the hippocampus of young, three-month-old transgenic McGill-Thy1-APP mice in the absence of amyloid plaques but associated with intracellular accumulation of amyloid beta petide oligomers. The role of such a pro-inflammatory process in the progression of the pathology remained to be elucidated.Methods and results: To clarify this we administered minocycline, a tetracyclic derivative with anti-inflammatory and neuroprotective properties, to young, pre-plaque McGill-Thy1-APP mice for one month. The treatment ended at the age of three months, when the mice were still devoid of plaques. Minocycline treatment corrected the up-regulation of inducible nitric oxide synthase and cyclooxygenase-2 observed in young transgenic placebo mice. Furthermore, the down-regulation of inflammatory markers correlated with a reduction in amyloid precursor protein levels and amyloid precursor protein-related products. Beta-site amyloid precursor protein cleaving enzyme 1 activity and levels were found to be up-regulated in transgenic placebo mice, while minocycline treatment restored these levels to normality. The anti-inflammatory and beta-secretase 1 effects could be partly explained by the inhibition of the nuclear factor kappa B pathway.Conclusions: Our study suggests that the pharmacological modulation of neuroinflammation might represent a promising approach for preventing or delaying the development of Alzheimer's disease neuropathology at its initial, pre-clinical stages. The results open new vistas to the interplay between inflammation and amyloid pathology. © 2012 Ferretti et al; licensee BioMed Central Ltd.
dc.publisherBMC
dc.rightsAttribution 4.0 International
dc.rights.urihttp://creativecommons.org/licenses/by/4.0/
dc.sourceUnpaywall 20201031
dc.subjectamyloid precursor protein
dc.subjectbeta secretase 1
dc.subjectcyclooxygenase 2
dc.subjectimmunoglobulin enhancer binding protein
dc.subjectinducible nitric oxide synthase
dc.subjectm 9511
dc.subjectminocycline
dc.subjectplacebo
dc.subjectunclassified drug
dc.subjectactin binding protein
dc.subjectAif1 protein, mouse
dc.subjectamyloid beta protein
dc.subjectamyloid beta protein (1 42)
dc.subjectamyloid beta protein[1-40]
dc.subjectamyloid beta-protein (1-42)
dc.subjectamyloid precursor protein
dc.subjectaspartic proteinase
dc.subjectBace1 protein, mouse
dc.subjectcalcium binding protein
dc.subjectCTF 1 transcription factor
dc.subjectCTF-1 transcription factor
dc.subjectcyclooxygenase 2
dc.subjectinducible nitric oxide synthase
dc.subjectinterleukin 1beta
dc.subjectminocycline
dc.subjectnuclear factor I
dc.subjectpeptide fragment
dc.subjectPtgs2 protein, mouse
dc.subjectsecretase
dc.subjectAlzheimer disease
dc.subjectamyloid plaque
dc.subjectanimal cell
dc.subjectanimal experiment
dc.subjectanimal model
dc.subjectanimal tissue
dc.subjectantiinflammatory activity
dc.subjectarticle
dc.subjectcontrolled study
dc.subjectdown regulation
dc.subjectdrug effect
dc.subjectdrug mechanism
dc.subjectenzyme activity
dc.subjectenzyme induction
dc.subjectfemale
dc.subjectmale
dc.subjectmouse
dc.subjectnervous system inflammation
dc.subjectnonhuman
dc.subjecttransgenic mouse
dc.subjectAlzheimer disease
dc.subjectamyloid neuropathy
dc.subjectanalysis of variance
dc.subjectanimal
dc.subjectC57BL mouse
dc.subjectdisease model
dc.subjectdown regulation
dc.subjectenzyme linked immunosorbent assay
dc.subjectgenetics
dc.subjecthuman
dc.subjectmetabolism
dc.subjectmutation
dc.subjectneurogenic inflammation
dc.subjectnewborn
dc.subjectAlzheimer Disease
dc.subjectAmyloid beta-Peptides
dc.subjectAmyloid beta-Protein Precursor
dc.subjectAmyloid Neuropathies
dc.subjectAmyloid Precursor Protein Secretases
dc.subjectAnalysis of Variance
dc.subjectAnimals
dc.subjectAnimals, Newborn
dc.subjectAspartic Acid Endopeptidases
dc.subjectCalcium-Binding Proteins
dc.subjectCyclooxygenase 2
dc.subjectDisease Models, Animal
dc.subjectDown-Regulation
dc.subjectEnzyme-Linked Immunosorbent Assay
dc.subjectHumans
dc.subjectInterleukin-1beta
dc.subjectMice
dc.subjectMice, Inbred C57BL
dc.subjectMice, Transgenic
dc.subjectMicrofilament Proteins
dc.subjectMinocycline
dc.subjectMutation
dc.subjectNeurogenic Inflammation
dc.subjectNFI Transcription Factors
dc.subjectNitric Oxide Synthase Type II
dc.subjectPeptide Fragments
dc.typeArticle
dc.contributor.departmentPHARMACOLOGY
dc.description.doi10.1186/1742-2094-9-62
dc.description.sourcetitleJournal of Neuroinflammation
dc.description.volume9
dc.description.page62
dc.published.statepublished
Appears in Collections:Staff Publications
Elements

Show simple item record
Files in This Item:
File Description SizeFormatAccess SettingsVersion 
10_1186_1742-2094-9-62.pdf591.24 kBAdobe PDF

OPEN

NoneView/Download

Google ScholarTM

Check

Altmetric


This item is licensed under a Creative Commons License Creative Commons