Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 22(15)2021 Jul 30.
Article in English | MEDLINE | ID: mdl-34360951

ABSTRACT

Epidemiological studies have implied that the nonsteroidal anti-inflammatory drug (NSAID) indomethacin slows the development and progression of Alzheimer's disease (AD). However, the underlying mechanisms are notably understudied. Using a chimeric mouse/human amyloid precursor protein (Mo/HuAPP695swe) and a mutant human presenilin 1 (PS1-dE9) (APP/PS1) expressing transgenic (Tg) mice and neuroblastoma (N) 2a cells as in vivo and in vitro models, we revealed the mechanisms of indomethacin in ameliorating the cognitive decline of AD. By screening AD-associated genes, we observed that a marked increase in the expression of α2-macroglobulin (A2M) was markedly induced after treatment with indomethacin. Mechanistically, upregulation of A2M was caused by the inhibition of cyclooxygenase-2 (COX-2) and lipocalin-type prostaglandin D synthase (L-PGDS), which are responsible for the synthesis of prostaglandin (PG)H2 and PGD2, respectively. The reduction in PGD2 levels induced by indomethacin alleviated the suppression of A2M expression through a PGD2 receptor 2 (CRTH2)-dependent mechanism. Highly activated A2M not only disrupted the production and aggregation of ß-amyloid protein (Aß) but also induced Aß efflux from the brain. More interestingly, indomethacin decreased the degradation of the A2M receptor, low-density lipoprotein receptor-related protein 1 (LRP1), which facilitated the brain efflux of Aß. Through the aforementioned mechanisms, indomethacin ameliorated cognitive decline in APP/PS1 Tg mice by decreasing Aß production and clearing Aß from the brains of AD mice.


Subject(s)
Amyloid beta-Peptides/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Indomethacin/pharmacology , Plaque, Amyloid/drug therapy , alpha-Macroglobulins/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Brain/drug effects , Brain/metabolism , Cell Line, Tumor , Cyclooxygenase 2/metabolism , Humans , Indomethacin/therapeutic use , Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Mice , Mice, Inbred C57BL , Plaque, Amyloid/metabolism , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Up-Regulation , alpha-Macroglobulins/genetics
2.
Mol Neurobiol ; 58(3): 1114-1127, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33095414

ABSTRACT

Prostaglandin (PG) A1 is a metabolic product of cyclooxygenase 2 (COX-2) that is potentially involved in regulating the development and progression of Alzheimer's disease (AD). PGA1 is a cyclopentenone (cy) PG characterized by the presence of a chemically reactive α,ß-unsaturated carbonyl. PGA1 is potentially involved in the regulation of multiple biological processes via Michael addition; however, the specific roles of PGA1 in AD remain unclear. TauP301S transgenic (Tg) mice were used as in vivo AD models, and neuroblastoma (N) 2a cells were used as an in vitro neuronal model. The PGA1-binding proteins were identified by HPLC-MS-MS after intracerebroventricular injection (i.c.v) of PGA1. Western blotting was used to determine tau phosphorylation in PGA1-treated Tg mice in the absence or in the presence of okadaic acid (OA), an inhibitor of protein phosphatase (PP) 2A. A combination of pull-down assay, immunoprecipitation, western blotting, and HPLC-MS-MS was used to determine that the PP2A scaffold subunit A alpha (PPP2R1A) is activated by the direct binding of PGA1 to cysteine 377. The effect of inhibiting tau hyperphosphorylation was tested in the Morris maze to determine the inhibitory effects of PGA1 on cognitive decline in tauP301S Tg mice. Incubation with N2a cells, pull-down assay, and mass spectrometry (MS) analysis revealed and indicated that PGA1 binds to more than 1000 proteins; some of these proteins are associated with AD and especially with tauopathies. Moreover, short-term administration of PGA1 in tauP301S Tg mice significantly decreased tau phosphorylation at Thr181, Ser202, and Ser404 in a dose-dependent manner. This effect was caused by the activation of PPP2R1A in tauP301S Tg mice. Importantly, PGA1 can form a Michael adduct with cysteine 377 of PPP2R1A, which is critical for the enzymatic activity of PP2A. Long-term treatment of tauP301S Tg mice with PGA1 activated PP2A and significantly reduced tau phosphorylation resulting in improvements in cognitive decline in tauP301S Tg mice. Our data provided new insight into the mechanisms of the ameliorating effects of PGA1 on cognitive decline in tauP301S Tg mice by activating PP2A via a mechanism involving the formation of a Michael adduct with cysteine 377 of PPP2R1A.


Subject(s)
Cysteine/metabolism , Prostaglandins A/pharmacology , Protein Phosphatase 2/metabolism , tau Proteins/metabolism , Animals , Cell Line, Tumor , Cognitive Dysfunction/pathology , HEK293 Cells , Humans , Mice, Transgenic , Phosphorylation/drug effects , Prostaglandins A/administration & dosage , Protein Subunits/metabolism
3.
Neurotherapeutics ; 16(2): 505-522, 2019 04.
Article in English | MEDLINE | ID: mdl-30627958

ABSTRACT

Prostaglandins (PGs) are early and key contributors to chronic neurodegenerative diseases. As one important member of classical PGs, PGA1 has been reported to exert potential neuroprotective effects. However, the mechanisms remain unknown. To this end, we are prompted to investigate whether PGA1 is a useful neurological treatment for Alzheimer's disease (AD) or not. Using high-throughput sequencing, we found that PGA1 potentially regulates cholesterol metabolism and lipid transport. Interestingly, we further found that short-term administration of PGA1 decreased the levels of the monomeric and oligomeric ß-amyloid protein (oAß) in a cholesterol-dependent manner. In detail, PGA1 activated the peroxisome proliferator-activated receptor-gamma (PPARγ) and ATP-binding cassette subfamily A member 1 (ABCA1) signalling pathways, promoting the efflux of cholesterol and decreasing the intracellular cholesterol levels. Through PPARγ/ABCA1/cholesterol-dependent pathway, PGA1 decreased the expression of presenilin enhancer protein 2 (PEN-2), which is responsible for the production of Aß. More importantly, long-term administration of PGA1 remarkably decreased the formation of Aß monomers, oligomers, and fibrils. The actions of PGA1 on the production and deposition of Aß ultimately improved the cognitive decline of the amyloid precursor protein/presenilin1 (APP/PS1) transgenic mice.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Cholesterol/metabolism , Cognition/drug effects , Cognitive Dysfunction/drug therapy , PPAR gamma/metabolism , Prostaglandins A/pharmacology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cognitive Dysfunction/metabolism , Mice , Mice, Transgenic , Presenilin-1/genetics , Presenilin-1/metabolism , Prostaglandins A/therapeutic use , Signal Transduction/drug effects
4.
Aging Cell ; 15(5): 861-71, 2016 10.
Article in English | MEDLINE | ID: mdl-27240539

ABSTRACT

Cyclooxygenase-2 (COX-2) has been recently identified to be involved in the pathogenesis of Alzheimer's disease (AD). Yet, the role of an important COX-2 metabolic product, prostaglandin (PG) I2 , in the pathogenesis of AD remains unknown. Using human- and mouse-derived neuronal cells as well as amyloid precursor protein/presenilin 1 (APP/PS1) transgenic mice as model systems, we elucidated the mechanism of anterior pharynx-defective (APH)-1α and pharynx-defective-1ß induction. In particular, we found that PGI2 production increased during the course of AD development. Then, PGI2 accumulation in neuronal cells activates PKA/CREB and JNK/c-Jun signaling pathways by phosphorylation, which results in APH-1α/1ß expression. As PGI2 is an important metabolic by-product of COX-2, its suppression by NS398 treatment decreases the expression of APH-1α/1ß in neuronal cells and APP/PS1 mice. More importantly, ß-amyloid protein (Aß) oligomers in the cerebrospinal fluid (CSF) of APP/PS1 mice are critical for stimulating the expression of APH-1α/1ß, which was blocked by NS398 incubation. Finally, the induction of APH-1α/1ß was confirmed in the brains of patients with AD. Thus, these findings not only provide novel insights into the mechanism of PGI2 -induced AD progression but also are instrumental for improving clinical therapies to combat AD.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Endopeptidases/genetics , Epoprostenol/pharmacology , Presenilin-1/metabolism , Up-Regulation/drug effects , Administration, Intranasal , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/cerebrospinal fluid , Animals , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Endopeptidases/metabolism , Female , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Proteins , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Nitrobenzenes/administration & dosage , Nitrobenzenes/pharmacology , Signal Transduction/drug effects , Sulfonamides/administration & dosage , Sulfonamides/pharmacology
5.
FASEB J ; 29(12): 5044-58, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26293690

ABSTRACT

Alzheimer's disease (AD) is associated with a magnesium ion (Mg(2+)) deficit in the serum or brain. However, the mechanisms regulating the roles of Mg(2+) in the pathologic condition of AD remain unknown. We studied whether brain Mg(2+) can decrease ß-amyloid (Aß) deposition and ameliorate the cognitive decline in a model of AD, the APPswe/PS1DE9 transgenic (Tg) mouse. We used a recently developed compound, magnesium-L-threonate (MgT), for a treatment that resulted in enhanced clearance of Aß in an anterior pharynx-defective (APH)-1α/-1ß-dependent manner. To further explore how MgT treatment inhibits cognitive decline in APP/PS1 Tg mice, the critical molecules for amyloid precursor protein (APP) cleavage and signaling pathways were investigated. In neurons, ERK1/2 and PPARγ signaling pathways were activated by MgT treatment, which in turn suppressed (by >80%) the expression of APH-1α/-1ß, which is responsible for the deposition of Aß and potentially contributes to the memory deficit that occurs in AD. More important, Aß oligomers in the cerebrospinal fluid (CSF) further promoted the expression of APH-1α/-1ß (by >2.5-fold), which enhances the γ-cleavage of APP and Aß deposition during AD progression. These findings provide new insights into the mechanisms of AD progression and are instrumental for developing better strategies to combat the disease.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Cognition Disorders/prevention & control , Endopeptidases/metabolism , Magnesium/pharmacology , Presenilin-1/genetics , Animals , Cell Line, Tumor , Humans , Male , Maze Learning , Membrane Proteins , Mice , Mice, Transgenic
6.
Anal Biochem ; 467: 40-6, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25205651

ABSTRACT

Using an ultrasensitive chemically reduced graphene oxide and ionic liquid modified glass carbon electrode (RGI-GCE), separated electrochemical signals of adenine and hypoxanthine in both human breast cancer (MCF-7) and mouse embryonic fibroblast (BALB/3T3) cells were observed. For the first time, low-level expression of purine bases in noncancerous BALB/3T3 cells can be electrochemically monitored. The metabolism of purine bases in carcinogen agent-contaminated BALB/3T3 cells was also investigated through the change of electrochemical signals ascribed to different purine bases, which opens a new electrochemical approach to the exploration of a low-level purine mechanism in noncancerous cells.


Subject(s)
Adenine/analysis , Carbon/chemistry , Electrochemical Techniques/methods , Electrodes , Glass/chemistry , Graphite/chemistry , Hypoxanthine/analysis , Adenine/metabolism , Animals , BALB 3T3 Cells , Biosensing Techniques/methods , Humans , Hypoxanthine/metabolism , MCF-7 Cells , Mice , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL
...