Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 219
Filter
Add more filters

Publication year range
1.
Int J Neuropsychopharmacol ; 26(9): 585-598, 2023 09 25.
Article in English | MEDLINE | ID: mdl-37490542

ABSTRACT

BACKGROUND: Alzheimer disease (AD) and depression often cooccur, and inhibition of phosphodiesterase-4 (PDE4) has been shown to ameliorate neurodegenerative illness. Therefore, we explored whether PDE4 inhibitor rolipram might also improve the symptoms of comorbid AD and depression. METHODS: APP/PS1/tau mice (10 months old) were treated with or without daily i.p. injections of rolipram for 10 days. The animal groups were compared in behavioral tests related to learning, memory, anxiety, and depression. Neurochemical measures were conducted to explore the underlying mechanism of rolipram. RESULTS: Rolipram attenuated cognitive decline as well as anxiety- and depression-like behaviors. These benefits were attributed at least partly to the downregulation of amyloid-ß, Amyloid precursor protein (APP), and Presenilin 1 (PS1); lower tau phosphorylation; greater neuronal survival; and normalized glial cell function following rolipram treatment. In addition, rolipram upregulated B-cell lymphoma-2 (Bcl-2) and downregulated Bcl-2-associated X protein (Bax) to reduce apoptosis; it also downregulated interleukin-1ß, interleukin-6, and tumor necrosis factor-α to restrain neuroinflammation. Furthermore, rolipram increased cAMP, PKA, 26S proteasome, EPAC2, and phosphorylation of ERK1/2 while decreasing EPAC1. CONCLUSIONS: Rolipram may mitigate cognitive deficits and depression-like behavior by reducing amyloid-ß pathology, tau phosphorylation, neuroinflammation, and apoptosis. These effects may be mediated by stimulating cAMP/PKA/26S and cAMP/exchange protein directly activated by cAMP (EPAC)/ERK signaling pathways. This study suggests that PDE4 inhibitor rolipram can be an effective target for treatment of comorbid AD and depression.


Subject(s)
Alzheimer Disease , Phosphodiesterase 4 Inhibitors , Mice , Animals , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/pharmacology , Rolipram/pharmacology , Mice, Transgenic , Phosphodiesterase 4 Inhibitors/pharmacology , Neuroinflammatory Diseases , Presenilin-1/metabolism , Presenilin-1/pharmacology , Depression/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Memory Disorders/drug therapy , Apoptosis , Disease Models, Animal
2.
Behav Brain Funct ; 19(1): 7, 2023 Apr 13.
Article in English | MEDLINE | ID: mdl-37055801

ABSTRACT

Increasing evidence has shown that the NOD-like receptor protein 1 (NLRP1) inflammasome is associated with Aß generation and deposition, which contributes to neuronal damage and neuronal-inflammation in Alzheimer's disease (AD). However, the specific mechanism of NLRP1 inflammasome in the pathogenesis of AD is still unclear. It has been reported that autophagy dysfunction can aggravate the pathological symptoms of AD and plays an important role in regulating Aß generation and clearance. We hypothesized that NLRP1 inflammasome activation may induce autophagy dysfunction contributing to the progression of AD. In the present study, we observed the relationship between Aß generation and NLRP1 inflammasome activation, as well as AMPK/mTOR mediated-autophagy dysfunction in WT 9-month-old (M) mice, APP/PS1 6 M and APP/PS1 9 M mice. Additionally, we further studied the effect of NLRP1 knockdown on cognitive function, Aß generation, neuroinflammation and AMPK/mTOR mediated autophagy in APP/PS1 9 M mice. Our results indicated that NLRP1 inflammasome activation and AMPK/mTOR mediated-autophagy dysfunction are closely implicated in Aß generation and deposition in APP/PS1 9 M mice, but not in APP/PS1 6 M mice. Meanwhile, we found that knockdown of NLRP1 significantly improved learning and memory impairments, decreased the expressions of NLRP1, ASC, caspase-1, p-NF-κB, IL-1ß, APP, CTF-ß, BACE1 and Aß1-42, and decreased the level of p-AMPK, Beclin 1 and LC3 II, and increased the level of p-mTOR and P62 in APP/PS1 9 M mice. Our study suggested that inhibition of NLRP1 inflammasome activation improves AMPK/mTOR mediated-autophagy dysfunction, resulting in the decrease of Aß generation, and NLRP1 and autophagy might be important targets to delay the progression of AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Mice , Animals , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/pharmacology , Inflammasomes/metabolism , Inflammasomes/pharmacology , Amyloid Precursor Protein Secretases/pharmacology , NLR Proteins , AMP-Activated Protein Kinases/pharmacology , Mice, Transgenic , Aspartic Acid Endopeptidases/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Autophagy , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/pharmacology , Disease Models, Animal
3.
J Asian Nat Prod Res ; 25(4): 387-402, 2023 Apr.
Article in English | MEDLINE | ID: mdl-35672874

ABSTRACT

The aim of this study is to explore the effect and mechanism of 3,6'-disinapoylsucrose (DISS) on an Alzheimer's disease (AD) mice model induced by APPswe695 lentivirus (LV) and intraperitoneal injection of lipopolysaccharide (LPS). The results show that DISS improves cognitive ability, decreases the levels of IL-2, IL-6, IL-1ß, and TNF-α, reduces the expression of NF-κB p65, and alleviates Aß deposition and nerve cell damage. DISS can regulate tyrosine kinase B (TrkB)/brain-derived neurotrophic factor (BDNF) signaling in the hippocampus. In summary, DISS can significantly alleviate neuroinflammation, spatial learning and memory disorders in AD model mice.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Mice , Animals , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/pharmacology , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Lipopolysaccharides/pharmacology , Up-Regulation , Cognitive Dysfunction/metabolism , Alzheimer Disease/metabolism , Hippocampus/metabolism , Amyloid beta-Peptides/metabolism
4.
Neuromodulation ; 26(3): 589-600, 2023 Apr.
Article in English | MEDLINE | ID: mdl-35595603

ABSTRACT

OBJECTIVES: Transauricular vagal nerve stimulation (taVNS) at 40 Hz attenuates hippocampal amyloid load in 6-month-old amyloid precursor protein/presenilin 1 (APP/PS1) transgenic mice, but it is unclear whether 40-Hz taVNS can improve cognition in these mice. Moreover, the underlying mechanisms are still unclear. MATERIALS AND METHODS: 6-month-old C57BL/6 (wild type [WT]) and APP/PS1 mice were subjected to 40-Hz taVNS. Novel Object Recognition and the Morris Water Maze were used to evaluate cognition. Hippocampal amyloid-ß (Aß)1-40, Aß1-42, pro-interleukin (IL)-1ß, and pro-IL-18 were measured using enzyme-linked immunosorbent assays. Hippocampal Aß42, purinergic 2X7 receptor (P2X7R), nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3 (NLRP3), Caspase-1, IL-1ß, and IL-18 expression were evaluated by western blotting. Histologic assessments including immunofluorescence, immunohistochemistry, Nissl staining, and Congo red staining were used to assess microglial phagocytosis, neuroprotective effects, and Aß plaque load. RESULTS: 40-Hz taVNS improved spatial memory and learning in 6-month-old APP/PS1 mice but did not affect recognition memory. There were no effects on the cognitive behaviors of 6-month-old WT mice. taVNS at 40 Hz modulated microglia; significantly decreased levels of Aß1-40, Aß1-42, pro-IL-1ß, and pro-IL-18; inhibited Aß42, P2X7R, NLRP3, Caspase-1, IL-1ß, and IL-18 expression; reduced Aß deposits; and had neuroprotective effects in the hippocampus of 6-month-old APP/PS1 mice. These changes were not observed in 6-month-old WT mice. CONCLUSION: Our results show that 40-Hz taVNS inhibits the hippocampal P2X7R/NLRP3/Caspase-1 signaling and improves spatial learning and memory in 6-month-old APP/PS1 mice.


Subject(s)
Neuroprotective Agents , Vagus Nerve Stimulation , Mice , Animals , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Interleukin-18/metabolism , Interleukin-18/pharmacology , Spatial Learning , Presenilin-1/genetics , Presenilin-1/metabolism , Presenilin-1/pharmacology , Caspase 1/metabolism , Caspase 1/pharmacology , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Mice, Inbred C57BL , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Mice, Transgenic , Hippocampus/metabolism
5.
J Nutr ; 152(1): 140-152, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34636875

ABSTRACT

BACKGROUND: There is growing evidence of strong associations between the pathogenesis of Alzheimer's disease (AD) and dysbiotic oral and gut microbiota. Recent studies demonstrated that isoorientin (ISO) is anti-inflammatory and alleviates markers of AD, which were hypothesized to be mediated by the oral and gut microbiota. OBJECTIVES: We studied the effects of oral administration of ISO on AD-related markers and the oral and gut microbiota in mice. METHODS: Eight-month-old amyloid precursor protein/presenilin-1 (AP) transgenic male mice were randomly allocated to 3 groups of 15 mice each: vehicle (AP) alone or with a low dose of ISO (AP + ISO-L; 25 mg/kg) or a high dose of ISO (AP + ISO-H; 50 mg/kg). Age-matched wild-type (WT) C57BL/6 male littermates were used as controls. The 4 groups were treated intragastrically with ISO or sterilized ultrapure water for 2 months. AD-related markers in the brain, serum, colon, and liver were analyzed with immunohistochemical and histochemical staining, Western blotting, and ELISA. Oral and gut microbiotas were analyzed using 16S ribosomal RNA gene sequencing. RESULTS: The high-dose ISO treatment significantly decreased amyloid beta 42-positive deposition by 38.1% and 45.2% in the cortex and hippocampus, respectively, of AP mice (P < 0.05). Compared with the AP group, both ISO treatments reduced brain phospho-Tau, phosphor-p65, phosphor-inhibitor of NF-κB, and brain and serum LPS and TNF-α by 17.9%-72.5% and increased brain and serum IL-4 and IL-10 by 130%-210% in the AP + ISO-L and AP + ISO-H groups (P < 0.05). Abundances of 26, 25, and 23 microbial taxa in oral, fecal and cecal samples, respectively, were increased in both the AP + ISO-L and AP + ISO-H groups relative to the AP group [linear discriminant analysis (LDA) >3.0; P < 0.05]. Gram-negative bacteria, Alteromonas, Campylobacterales, and uncultured Bacteroidales bacterium were positively correlated (rho = 0.28-0.59; P < 0.05) with the LPS levels and responses of inflammatory cytokines. CONCLUSIONS: The microbiota-gut-brain axis is a potential mechanism by which ISO reduces AD-related markers in AP mice.


Subject(s)
Alzheimer Disease , Gastrointestinal Microbiome , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/pharmacology , Amyloid beta-Protein Precursor/therapeutic use , Animals , Disease Models, Animal , Luteolin , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1
6.
Phytother Res ; 36(3): 1297-1309, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35088915

ABSTRACT

Enhancing glucagon-like peptide 1 (GLP-1) signaling with a dipeptidyl peptidase IV (DPP-4) inhibitor might exert protective effects on Alzheimer's disease (AD). We found that intragastric administration of Gramcyclin A (10, 20 and 40 mg/kg), a novel DPP-4 inhibitor, for 3 months significantly reversed cognitive decline in APP/PS1/tau triple transgenic mice in a dose-dependent manner. Gramcyclin A treatment markedly reduced Aß plaques as well as the insoluble and soluble forms of Aß40 and Aß42 in the hippocampus of APP/PS1/tau mice. Treatment with Gramcyclin A remarkedly decreased the level of microglia and suppressed neuroinflammation in the hippocampus of APP/PS1/tau mice. Moreover, Gramcyclin A treatment could increase brain glucose uptake in APP/PS1/tau mice, as detected by 18-fluoro-2-deoxyglucose (18 F-FDG) micro-positron emission tomography (micro-PET) imaging. Furthermore, Gramcyclin A significantly increased expression of glucagon-like peptide-1 (GLP-1), GLP-1R, proliferator-activated receptor gamma coactivator (PGC)-1α and glucose transporter 4 (GLUT4), and inhibited insulin receptor (IRS)-1 phosphorylation and tau hyperphosphorylation in the hippocampus of APP/PS1/tau mice. Collectively, Gramcyclin A conferred protective effects against AD via enhancing brain GLP-1-dependent glucose uptake. The DPP-4 inhibitor Gramcyclin A might be a potential therapeutic drug for AD.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Dipeptidyl-Peptidase IV Inhibitors , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/pharmacology , Animals , Brain , Cognition , Cognitive Dysfunction/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Disease Models, Animal , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide 1/therapeutic use , Glucose/metabolism , Hippocampus , Mice , Mice, Transgenic
7.
Cell Physiol Biochem ; 55(S3): 157-170, 2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34318654

ABSTRACT

BACKGROUND/AIMS: The Amyloid Precursor Protein (APP) is involved in the regulation of multiple cellular functions via protein-protein interactions and has been most studied with respect to Alzheimer's disease (AD). Abnormal processing of the single transmembrane-spanning C99 fragment of APP contributes to the formation of amyloid plaques, which are causally related to AD. Pathological C99 accumulation is thought to associate with early cognitive defects in AD. Here, unexpectedly, sequence analysis revealed that C99 exhibits 24% sequence identity with the KCNE1 voltage-gated potassium (Kv) channel ß subunit, comparable to the identity between KCNE1 and KCNE2-5 (21-30%). This suggested the possibility of C99 regulating Kv channels. METHODS: We quantified the effects of C99 on Kv channel function, using electrophysiological analysis of subunits expressed in Xenopus laevis oocytes, biochemical and immunofluorescence techniques. RESULTS: C99 isoform-selectively inhibited (by 30-80%) activity of a range of Kv channels. Among the KCNQ (Kv7) family, C99 isoform-selectively inhibited, shifted the voltage dependence and/or slowed activation of KCNQ2, KCNQ3, KCNQ2/3 and KCNQ5, with no effects on KCNQ1, KCNQ1-KCNE1 or KCNQ4. C99/APP co-localized with KCNQ2 and KCNQ3 in adult rat sciatic nerve nodes of Ranvier. Both C99 and full-length APP co-immunoprecipitated with KCNQ2 in vitro, yet unlike C99, APP only weakly affected KCNQ2/3 activity. Finally, C99 altered the effects on KCNQ2/3 function of inhibitors tetraethylammounium and XE991, but not openers retigabine and ICA27243. CONCLUSION: Our findings raise the possibility of C99 accumulation early in AD altering cellular excitability by modulating Kv channel activity.


Subject(s)
Amyloid beta-Protein Precursor/pharmacology , KCNQ Potassium Channels/genetics , KCNQ2 Potassium Channel/genetics , KCNQ3 Potassium Channel/genetics , Peptide Fragments/pharmacology , Amino Acid Sequence , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Anthracenes/pharmacology , Gene Expression , Humans , KCNQ Potassium Channels/metabolism , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Oocytes/cytology , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Peptide Fragments/genetics , Peptide Fragments/metabolism , Ranvier's Nodes/drug effects , Ranvier's Nodes/metabolism , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Tetraethylammonium/pharmacology , Xenopus laevis
8.
Exp Cell Res ; 396(1): 112266, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32905804

ABSTRACT

The aggregation of ß-amyloid (Aß) peptide in Alzheimer's disease (AD) is characterized by mitochondrial dysfunction and mitophagy impairment. Mitophagy is a homeostatic mechanism by which autophagy selectively eliminates damaged mitochondria. Valinomycin is a respiratory chain inhibitor that activates mitophagy via the PINK1/Parkin signaling pathway. However, the mechanism underlying the association between mitophagy and valinomycin in Aß formation has not been explored. Here, we demonstrate that genetically modified (N2a/APP695swe) cells overexpressing a mutant amyloid precursor protein (APP) serve as an in vitro model of AD for studying mitophagy and ATP-related metabolomics. Our results prove that valinomycin induced a time-dependent increase in the mitophagy activation of N2a/APP695swe cells as indicated by increased levels of PINK1, Parkin, and LC3II as well as increased the colocalization of Parkin-Tom20 and fewer mitochondria (indicated by decreased Tom20 levels). Valinomycin significantly decreased Aß1-42 and Aß1-40 levels after 3 h of treatment. ATP levels and ATP-related metabolites were significantly increased at this time. Our findings suggest that the elimination of impaired mitochondria via valinomycin-induced mitophagy ameliorates AD by decreasing Aß and improving ATP levels.


Subject(s)
Adenosine Triphosphate/biosynthesis , Amyloid beta-Peptides/genetics , Mitochondria/metabolism , Mitophagy/genetics , Peptide Fragments/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/pharmacology , Animals , Cell Line, Tumor , Gene Expression Regulation , Humans , Ionophores/pharmacology , Membrane Potential, Mitochondrial/drug effects , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Metabolomics/methods , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Precursor Protein Import Complex Proteins , Mitophagy/drug effects , Models, Biological , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Peptide Fragments/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Valinomycin/pharmacology
9.
J Neurosci ; 39(17): 3188-3203, 2019 04 24.
Article in English | MEDLINE | ID: mdl-30804097

ABSTRACT

Secreted amyloid precursor protein-alpha (sAPPα) has growth factor-like properties and can modulate long-term potentiation (LTP) and memory. Here, we demonstrate that exposure to sAPPα converts short-lasting LTP into protein-synthesis-dependent late LTP in hippocampal slices from male rats. sAPPß had no discernable effect. We hypothesized that sAPPα facilitated LTP via regulated glutamate receptor trafficking and de novo protein synthesis. We found using a linear mixed model that sAPPα stimulated trafficking of GluA2-lacking AMPARs, as well as NMDARs to the extrasynaptic cell surface, in a calcium/calmodulin-dependent kinase II and protein kinase G-dependent manner. Both cell surface receptor accumulation and LTP facilitation were present even after sAPPα washout and inhibition of receptor trafficking or protein synthesis prevented all these effects. Direct visualization of newly synthesized proteins (FUNCAT-PLA) confirmed the ability of sAPPα to stimulate de novo protein synthesis and revealed GluA1 as one of the upregulated proteins. Therefore, sAPPα generates a coordinated synthesis and trafficking of glutamate receptors to the cell surface that facilitate LTP.SIGNIFICANCE STATEMENT Secreted amyloid precursor protein-alpha (sAPPα) is a neurotrophic and neuroprotective protein that can promote synaptic plasticity and memory, yet the molecular mechanisms underlying these effects are still not well understood. Here, we show that sAPPα facilitates long-term potentiation (LTP) in a concentration-dependent fashion through cellular processes involving de novo protein synthesis and trafficking of both GluA2-lacking AMPARs and NMDARs to the extrasynaptic cell surface. sAPPα also enhances GluA1, but not GluA2, synthesis. The trafficking effects, along with the LTP facilitation, persist after sAPPα washout, revealing a metaplastic capability of exogenous sAPPα administration. sAPPα thus facilitates LTP through coordinated activation of protein synthesis and trafficking of glutamate receptors to the cell surface, where they are positioned for priming LTP.


Subject(s)
Amyloid beta-Protein Precursor/pharmacology , Hippocampus/physiology , Long-Term Potentiation/drug effects , Protein Biosynthesis/drug effects , Receptors, Glutamate/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Hippocampus/drug effects , Long-Term Potentiation/physiology , Male , Protein Biosynthesis/physiology , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Sprague-Dawley
10.
J Nat Prod ; 83(2): 223-230, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32031796

ABSTRACT

Cimicifuga dahurica has traditionally been used as an antipyretic, analgesic, and anti-inflammatory agent and as a treatment for uterine and anal prolapse. This study has investigated the potential beneficial effects of this medicinal plant and its components on Alzheimer's disease (AD) with a focus on amyloid beta (Aß) production and scopolamine-induced memory impairment in mice. An ethanol extract from C. dahurica roots decreased Aß production in APP-CHO cells [Chinese hamster ovarian (CHO) cells stably expressing amyloid precursor protein (APP)], as determined by an enzyme-linked immunosorbent assay and Western blot analysis. Then, the compounds isolated from C. dahurica were tested for their antiamyloidogenic activities. Four compounds (1-4) efficiently interrupted Aß generation by suppressing the level of ß-secretase in APP-CHO cells. Moreover, the in vivo experimental results demonstrated that compound 4 improved the cognitive performances of mice with scopolamine-induced disruption on behavioral tests and the expression of memory-related proteins. Taken together, these results suggest that C. dahurica and its constituents are potential agents for preventing or alleviating the symptoms of AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/pharmacology , Plants, Medicinal/chemistry , Scopolamine/pharmacology , Alzheimer Disease/diet therapy , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/pharmacology , Amyloid beta-Peptides/chemistry , Amyloid beta-Protein Precursor/metabolism , Animals , CHO Cells , Cimicifuga , Cricetinae , Cricetulus , Mice , Molecular Structure , Plants, Medicinal/metabolism , Scopolamine/metabolism
11.
J Biol Chem ; 293(33): 12663-12680, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29934309

ABSTRACT

Human tissue kallikrein (KLK) proteases are hormone-like signaling molecules with important functions in cancer pathophysiology. KLK-related peptidase 6 (KLK6), specifically, is highly up-regulated in several types of cancer, where its increased activity promotes cancer invasion and metastasis. This characteristic suggests KLK6 as an attractive target for therapeutic interventions. However, inhibitors that specifically target KLK6 have not yet been reported, possibly because KLK6 shares a high sequence homology and structural similarity with other serine proteases and resists inhibition by many polypeptide inhibitors. Here, we present an innovative combinatorial approach to engineering KLK6 inhibitors via flow cytometry-based screening of a yeast-displayed mutant library of the human amyloid precursor protein Kunitz protease inhibitor domain (APPI), an inhibitor of other serine proteases, such as anionic and cationic trypsins. On the basis of this screening, we generated APPIM17L,I18F,S19F,F34V (APPI-4M), an APPI variant with a KLK6 inhibition constant (Ki ) of 160 pm and a turnover time of 10 days. To the best of our knowledge, APPI-4M is the most potent KLK6 inhibitor reported to date, displaying 146-fold improved affinity and 13-fold improved proteolytic stability compared with WT APPI (APPIWT). We further demonstrate that APPI-4M acts as a functional inhibitor in a cell-based model of KLK6-dependent breast cancer invasion. Finally, the crystal structures of the APPIWT/KLK6 and APPI-4M/KLK6 complexes revealed the structural and mechanistic bases for the improved KLK6 binding and proteolytic resistance of APPI-4M. We anticipate that APPI-4M will have substantial translational potential as both imaging agent and therapeutic.


Subject(s)
Amyloid beta-Protein Precursor/pharmacology , Breast Neoplasms/drug therapy , Genetic Engineering , Kallikreins/antagonists & inhibitors , Protease Inhibitors/pharmacology , Proteolysis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement , Cell Proliferation , Female , High-Throughput Screening Assays , Humans , Kallikreins/chemistry , Models, Molecular , Protein Binding , Protein Conformation , Tumor Cells, Cultured
12.
FASEB J ; 31(10): 4434-4446, 2017 10.
Article in English | MEDLINE | ID: mdl-28646018

ABSTRACT

The amyloid precursor protein (APP) has long been appreciated for its role in Alzheimer's disease (AD) pathology. However, less is known about the physiologic function of APP outside of AD. Particularly, whether and how APP may regulate functions of cell surface receptors, including GPCRs, remains largely unclear. In this study, we identified a novel direct interaction between APP and the α2A-adrenergic receptor (α2AAR) that occurs at the intracellular domains of both proteins. The APP interaction with α2AAR is promoted by agonist stimulation and competes with arrestin 3 binding to the receptor. Consequently, the presence of APP attenuates α2AAR internalization and desensitization, which are arrestin-dependent processes. Furthermore, in neuroblastoma neuro-2A cells and primary superior cervical ganglion neurons, where APP is highly expressed, the lack of APP leads to a dramatic increase in plasma membrane recruitment of endogenous arrestin 3 following α2AAR activation. Concomitantly, agonist-induced internalization of α2AAR is significantly enhanced in these neuronal cells. Our study provided the first evidence that APP fine tunes GPCR signaling and trafficking. Given the important role of α2AAR in controlling norepinephrine release and response, this novel regulation of α2AAR by APP may have an impact on modulation of noradrenergic activity and sympathetic tone.-Zhang, F., Gannon, M., Chen, Y., Zhou, L., Jiao, K., Wang, Q. The amyloid precursor protein modulates α2A-adrenergic receptor endocytosis and signaling through disrupting arrestin 3 recruitment.


Subject(s)
Arrestins/metabolism , Endocytosis/drug effects , Neurons/metabolism , Receptors, Adrenergic, alpha-2/metabolism , beta-Arrestin 2/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/pharmacology , Animals , Endocytosis/physiology , Mice , Protein Transport/physiology , Signal Transduction/physiology
13.
J Neurosci ; 36(32): 8471-86, 2016 08 10.
Article in English | MEDLINE | ID: mdl-27511018

ABSTRACT

UNLABELLED: Prior work suggests that amyloid precursor protein (APP) can function as a proinflammatory receptor on immune cells, such as monocytes and microglia. Therefore, we hypothesized that APP serves this function in microglia during Alzheimer's disease. Although fibrillar amyloid ß (Aß)-stimulated cytokine secretion from both wild-type and APP knock-out (mAPP(-/-)) microglial cultures, oligomeric Aß was unable to stimulate increased secretion from mAPP(-/-) cells. This was consistent with an ability of oligomeric Aß to bind APP. Similarly, intracerebroventricular infusions of oligomeric Aß produced less microgliosis in mAPP(-/-) mice compared with wild-type mice. The mAPP(-/-) mice crossed to an APP/PS1 transgenic mouse line demonstrated reduced microgliosis and cytokine levels and improved memory compared with wild-type mice despite robust fibrillar Aß plaque deposition. These data define a novel function for microglial APP in regulating their ability to acquire a proinflammatory phenotype during disease. SIGNIFICANCE STATEMENT: A hallmark of Alzheimer's disease (AD) brains is the accumulation of amyloid ß (Aß) peptide within plaques robustly invested with reactive microglia. This supports the notion that Aß stimulation of microglial activation is one source of brain inflammatory changes during disease. Aß is a cleavage product of the ubiquitously expressed amyloid precursor protein (APP) and is able to self-associate into a wide variety of differently sized and structurally distinct multimers. In this study, we demonstrate both in vitro and in vivo that nonfibrillar, oligomeric forms of Aß are able to interact with the parent APP protein to stimulate microglial activation. This provides a mechanism by which metabolism of APP results in possible autocrine or paracrine Aß production to drive the microgliosis associated with AD brains.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Microglia/metabolism , Adaptation, Ocular/genetics , Adaptation, Ocular/physiology , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/pharmacology , Animals , Astrocytes/metabolism , Cell Proliferation/genetics , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Exploratory Behavior/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morpholinos/pharmacology , Mutation/genetics , Phenotype , Presenilin-1/genetics , Presenilin-1/metabolism
14.
Neurobiol Learn Mem ; 138: 291-299, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27521248

ABSTRACT

Secreted amyloid precursor protein-α (sAPPα) is a neurotrophic and neuroprotective molecule which can enhance learning and synaptic plasticity. Aging is associated with memory decline and impaired long-term potentiation (LTP). SAPPα therefore has potential as a nootropic agent which could be used to offset age-related cognitive decline. In this study we investigated the effects of sAPPα on spatial memory tasks and LTP in aged and young Long-Evans rats. Two hippocampus-dependent tasks were employed to measure spatial memory that is susceptible to impairments during aging. Aged rats showed a mild deficit in the novel object location task, but memory was significantly enhanced by bilateral intrahippocampal injections of sAPPα. There was no effect on the performance of young animals. In the watermaze task, however, sAPPα did not alleviate age-related decline in spatial memory. In subsequent electrophysiological experiments, LTP was impaired in slices from aged animals, but plasticity was rescued in a concentration-dependent manner by exogenous sAPPα administration. In contrast, LTP was impaired in young animals by sAPPα. Overall, these data support the hypothesis that sAPPα has therapeutic potential as a treatment for age-related cognitive decline.


Subject(s)
Aging/physiology , Amyloid beta-Protein Precursor/pharmacology , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Spatial Memory/drug effects , Animals , Dose-Response Relationship, Drug , Hippocampus/physiology , Long-Term Potentiation/physiology , Rats , Rats, Long-Evans , Spatial Memory/physiology
15.
J Neurochem ; 135(3): 630-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26342176

ABSTRACT

We recently found that sAPPα decreases amyloid-beta generation by directly associating with ß-site amyloid precursor protein (APP)-converting enzyme 1 (BACE1), thereby modulating APP processing. Because inhibition of BACE1 decreases glycogen synthase kinase 3 beta (GSK3ß)-mediated Alzheimer's disease (AD)-like tau phosphorylation in AD patient-derived neurons, we determined whether sAPPα also reduces GSK3ß-mediated tau phosphorylation. We initially found increased levels of inhibitory phosphorylation of GSK3ß (Ser9) in primary neurons from sAPPα over-expressing mice. Further, recombinant human sAPPα evoked the same phenomenon in SH-SY5Y cells. Further, in SH-SY5Y cells over-expressing BACE1, and HeLa cells over-expressing human tau, sAPPα reduced GSK3ß activity and tau phosphorylation. Importantly, the reductions in GSK3ß activity and tau phosphorylation elicited by sAPPα were prevented by BACE1 but not γ-secretase inhibition. In accord, AD mice over-expressing human sAPPα had less GSK3ß activity and tau phosphorylation compared with controls. These results implicate a direct relationship between APP ß-processing and GSK3ß-mediated tau phosphorylation and further define the central role of sAPPα in APP autoregulation and AD pathogenesis.


Subject(s)
Amyloid beta-Protein Precursor/pharmacology , Glycogen Synthase Kinase 3/physiology , Signal Transduction/physiology , tau Proteins/antagonists & inhibitors , tau Proteins/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3 beta , Humans , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Phosphorylation/physiology , Signal Transduction/drug effects
16.
Neurol Sci ; 36(4): 593-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25380623

ABSTRACT

This study was performed to understand whether P165 improves learning and memory by restoring insulin action using a diabetes mellitus (DM) rat model. A total of 34 male Sprague-Dawley rats were randomly divided into four groups: control group (n = 8), DM group (n = 8), DM group treated with a low dose of P165 (n = 9), and DM group treated with a high dose of P165 (n = 9). After 8 weeks of treatment, the animals were killed and the expression of insulin signaling-related proteins was examined in the hippocampus by Western blot and immunohistochemical staining. Administration of P165 in diabetic rats did not induce a significant effect on the fasting blood glucose level. The expression of IR, IRS-1, AKT, p-CREB, and Bcl-2 proteins was significantly enhanced in the hippocampus in diabetic rats. Treatment of diabetic rats with P165 at both low and high doses significantly attenuated the expression levels of these proteins. Moreover, immunohistochemistry staining showed that IR, IRS-1, AKT, p-CREB, and Bcl-2 were abundantly expressed in the CA1 region of the hippocampus. The number of cells positively stained for the above proteins was significantly higher in diabetic tissues compared to control tissues, whereas P165 treatments induced a significant reduction in the expression of these proteins. The expression of IR, IRS-1, AKT, p-CREB, and Bcl-2 was enhanced in DM rats, and administration of P165 normalized the expression of these molecules, suggesting that P165 can improve impaired insulin signal transduction.


Subject(s)
Amyloid beta-Protein Precursor/therapeutic use , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Hypoglycemic Agents/therapeutic use , Insulin/metabolism , Peptide Fragments/therapeutic use , Signal Transduction/drug effects , Amyloid beta-Protein Precursor/pharmacology , Analysis of Variance , Animals , Brain/drug effects , Brain/metabolism , CREB-Binding Protein/metabolism , Diabetes Mellitus, Experimental/chemically induced , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Male , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Insulin/metabolism , Streptozocin/toxicity
17.
J Neurochem ; 129(5): 756-69, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24517464

ABSTRACT

The ß-amyloid precursor protein (APP) has been extensively studied for its role as the precursor of the ß-amyloid protein (Aß) of Alzheimer's disease. However, the normal function of APP remains largely unknown. This article reviews studies on the structure, expression and post-translational processing of APP, as well as studies on the effects of APP in vitro and in vivo. We conclude that the published data provide strong evidence that APP has a trophic function. APP is likely to be involved in neural stem cell development, neuronal survival, neurite outgrowth and neurorepair. However, the mechanisms by which APP exerts its actions remain to be elucidated. The available evidence suggests that APP interacts both intracellularly and extracellularly to regulate various signal transduction mechanisms. This article reviews studies on the structure, expression and post-translational processing of ß-amyloid precursor protein (APP), as well as studies on the effects of APP in vitro and in vivo. We conclude that the published data provide strong evidence that APP has a trophic function. APP is likely to be involved in neural stem cell development, neuronal survival, neurite outgrowth and neurorepair. However, the mechanisms by which APP exerts its actions remain to be elucidated. The available evidence suggests that APP interacts both intracellularly and extracellularly to regulate various signal transduction mechanisms.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/physiology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/pharmacology , Animals , Cell Differentiation/physiology , Cell Proliferation , Humans , Mice , Neural Stem Cells/physiology , Promoter Regions, Genetic/genetics
18.
Biochem Biophys Res Commun ; 447(3): 394-9, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24727450

ABSTRACT

Alzheimer's disease (AD) is one of the most common neurodegenerative diseases leading to dementia. Although cytotoxicity of amyloid ß peptides has been intensively studied within pathophysiology of AD, the physiological function of amyloid precursor protein (APP) still remains unclarified. We have shown previously that secreted APPα (sAPPα) is associated with glial differentiation of neural stem cells. To elucidate specific mechanisms underlying sAPPα-induced gliogenesis, we examined the potential involvement of bone morphogenic proteins (BMPs). BMPs are one of the factors involved in glial differentiation of neural progenitor cells. When expressions of BMP-2, -4, and -7 were examined, upregulation of BMP-4 expression was solely observed as a result of treatment with sAPPα in a time and dose-dependent manner. Furthermore, the treatment of sAPPα promoted phosphorylation of Smad1/5/8, a downstream signaling mediator of BMP receptors. Interestingly, N-terminal domain of APP (1-205) was sufficient to elevate BMP4 expression, resulting in an increase of glial fibrillary acidic protein (GFAP) expression and phosphorylation of Smad1/5/8. However, the application of APP neutralizing antibody and anti-BMP4 antibody significantly suppressed expression of BMP-4 as well as phosphorylation of Smad1/5/8. Thus, our results indicate that sAPPα-induced gliogenesis is in part mediated by the BMP-4 signaling pathway. We also observed upregulation of BMP-4 and phosphorylation of Smad1/5/8 in APP transgenic mice. It is imperative to unravel the mechanisms underlying the role of BMP-4 during APPα-induced glial differentiation in hope of providing novel prevention or treatment for AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/physiology , Bone Morphogenetic Protein 4/metabolism , Neural Stem Cells/cytology , Neurogenesis/physiology , Neuroglia/cytology , Peptide Fragments/physiology , Smad Proteins/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/pharmacology , Animals , Bone Morphogenetic Protein 4/genetics , Cell Line, Tumor , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Mice, Transgenic , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Phosphorylation , Signal Transduction , Up-Regulation
19.
J Neurosci Res ; 92(11): 1478-89, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24916405

ABSTRACT

The function of the ß-amyloid precursor protein (APP) of Alzheimer's disease is poorly understood. The secreted ectodomain fragment of APP (sAPPα) can be readily cleaved to produce a small N-terminal fragment (N-APP) that contains heparin-binding and metal-binding domains and that has been found to have biological activity. In the present study, we examined whether N-APP can bind to lipids. We found that N-APP binds selectively to phosphoinositides (PIPs) but poorly to most other lipids. Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2 )-rich microdomains were identified on the extracellular surface of neurons and glia in primary hippocampal cultures. N-APP bound to neurons and colocalized with PIPs on the cell surface. Furthermore, the binding of N-APP to neurons increased the level of cell-surface PI(4,5)P2 and phosphatidylinositol 3,4,5-trisphosphate. However, PIPs were not the principal cell-surface binding site for N-APP, because N-APP binding to neurons was not inhibited by a short-acyl-chain PIP analogue, and N-APP did not bind to glial cells which also possessed PI(4,5)P2 on the cell surface. The data are explained by a model in which N-APP binds to two distinct components on neurons, one of which is an unidentified receptor and the second of which is a PIP lipid, which binds more weakly to a distinct site within N-APP. Our data provide further support for the idea that N-APP may be an important mediator of APP's biological activity.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cell Membrane/metabolism , Hippocampus/cytology , Phosphatidylinositols/metabolism , Protein Binding/physiology , Amyloid beta-Protein Precursor/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Binding Sites/drug effects , Cell Membrane/drug effects , Cells, Cultured , Glial Fibrillary Acidic Protein/metabolism , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Phosphatidylinositol Phosphates/metabolism , Protein Binding/drug effects
20.
Blood ; 120(3): 671-7, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22674803

ABSTRACT

Coagulation factor XI (FXI) plays an important part in both venous and arterial thrombosis, rendering FXIa a potential target for the development of antithrombotic therapy. The kunitz protease inhibitor (KPI) domain of protease nexin-2 (PN2) is a potent, highly specific inhibitor of FXIa, suggesting its possible role in the inhibition of FXI-dependent thrombosis in vivo. Therefore, we examined the effect of PN2KPI on thrombosis in the murine carotid artery and the middle cerebral artery. Intravenous administration of PN2KPI prolonged the clotting time of both human and murine plasma, and PN2KPI inhibited FXIa activity in both human and murine plasma in vitro. The intravenous administration of PN2KPI into WT mice dramatically decreased the progress of FeCl(3)-induced thrombus formation in the carotid artery. After a similar initial rate of thrombus formation with and without PN2KPI treatment, the propagation of thrombus formation after 10 minutes and the amount of thrombus formed were significantly decreased in mice treated with PN2KPI injection compared with untreated mice. In the middle cerebral artery occlusion model, the volume and fraction of ischemic brain tissue were significantly decreased in PN2KPI-treated compared with untreated mice. Thus, inhibition of FXIa by PN2KPI is a promising approach to antithrombotic therapy.


Subject(s)
Amyloid beta-Protein Precursor/pharmacology , Carotid Artery Thrombosis/blood , Carotid Artery Thrombosis/drug therapy , Factor XIa/antagonists & inhibitors , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/drug therapy , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Animals , Anticoagulants/pharmacology , Bleeding Time , Blood Coagulation/drug effects , Blood Coagulation/physiology , Disease Models, Animal , Drug Design , Factor XIa/metabolism , Female , Humans , Injections, Intravenous , Mice , Mice, Inbred C57BL , Protein Structure, Tertiary/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL