Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 194
Filter
1.
Neurochem Res ; 46(11): 2832-2851, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34357520

ABSTRACT

Alzheimer's disease (AD) is one of the most prevailing neurodegenerative disorders of elderly humans associated with cognitive damage. Biochemical, epigenetic, and pathophysiological factors all consider a critical role of extracellular amyloid-beta (Aß) plaques and intracellular neurofibrillary tangles (NFTs) as pathological hallmarks of AD. In an endeavor to describe the intricacy and multifaceted nature of AD, several hypotheses based on the roles of Aß accumulation, tau hyperphosphorylation, impaired cholinergic signaling, neuroinflammation, and autophagy during the initiation and advancement of the disease have been suggested. However, in no way do these theories have the potential of autonomously describing the pathophysiological alterations located in AD. The complex pathological nature of AD has hindered the recognition and authentication of successful biomarkers for the progression of its diagnosis and therapeutic strategies. There has been a significant research effort to design multi-target-directed ligands for the treatment of AD, an approach which is developed by the knowledge that AD is a composite and multifaceted disease linked with several separate but integrated molecular pathways.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Anti-Inflammatory Agents/administration & dosage , Antibodies, Monoclonal/administration & dosage , tau Proteins/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Protein Precursor/antagonists & inhibitors , Amyloid beta-Protein Precursor/metabolism , Animals , Humans , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/therapy , Risk Reduction Behavior , tau Proteins/antagonists & inhibitors
2.
Neuropharmacology ; 197: 108737, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34343610

ABSTRACT

Alzheimer's disease (AD) is the most common dementia, and no disease-modifying therapeutic agents are currently available. BDNF/TrkB signaling is impaired in AD and is associated with prominent delta-secretase (δ-secretase, also known as asparaginyl endopeptidase or legumain) activation, which simultaneously cleaves both APP and Tau and promotes Aß production and neurofibrillary tangles (NFT) pathologies. Here we show that the optimized δ-secretase inhibitor (#11a) or TrkB receptor agonist (CF3CN) robustly blocks δ-secretase activity separately, and their combination synergistically blunts δ-secretase, exhibiting promising therapeutic efficacy in 3xTg AD mouse model. The optimal δ-secretase inhibitor reveals demonstrable brain exposure and oral bioavailability, suppressing APP N585 and Tau N368 cleavage by δ-secretase. Strikingly, CF3CN treatment evidently escalates BDNF levels. Both #11a and CF3CN display strong in vivo PK/PD properties and ability to suppress δ-secretase activity in the brain. Orally administrated CF3CN strongly activates TrkB that triggers active Akt to phosphorylate δ-secretase T322, preventing its proteolytic activation and mitigating AD pathologies. #11a or CF3CN significantly diminishes AD pathogenesis and improves cognitive functions with the combination exhibiting the maximal effect. Thus, our data support that these derivatives are strong pharmaceutical candidates for the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Brain-Derived Neurotrophic Factor/drug effects , Cysteine Endopeptidases/drug effects , Enzyme Inhibitors/therapeutic use , Membrane Glycoproteins/drug effects , Neuroprotective Agents/therapeutic use , Receptor, trkB/drug effects , Signal Transduction/drug effects , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/antagonists & inhibitors , Animals , Brain/drug effects , Brain/enzymology , Cognition/drug effects , Humans , Maze Learning/drug effects , Membrane Glycoproteins/agonists , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacokinetics , Rats , Receptor, trkB/agonists , tau Proteins/antagonists & inhibitors
3.
Adv Sci (Weinh) ; 8(11): e2003558, 2021 06.
Article in English | MEDLINE | ID: mdl-34105277

ABSTRACT

The binding of amyloid precursor protein (APP) expressed on tumor cells to death receptor 6 (DR6) could initiate the necroptosis pathway, which leads to necroptotic cell death of vascular endothelial cells (ECs) and results in tumor cells (TCs) extravasation and metastasis. This study reports the first inhibitor of DR6/APP interaction as a novel class of anti-hematogenous metastatic agent. By rationally utilizing three combined strategies including selection based on phage display library, d-retro-inverso modification, and multiple conjugation of screened peptidomimetic with 4-arm PEG, the polymer-peptidomimetic conjugate PEG-tAHP-DRI (tetra-(D-retro-inverso isomer of AHP-12) substitued 4-arm PEG5k ) is obtained as the most promising agent with the strongest binding potency (KD  = 51.12 × 10-9  m) and excellent pharmacokinetic properties. Importantly, PEG-tAHP-DRI provides efficient protection against TC-induced ECs necroptosis both in vitro and in vivo. Moreover, this ligand exhibits prominent anti-hematogenous metastatic activity in serval different metastatic mouse models (B16F10, 4T1, CT26, and spontaneous lung metastasis of 4T1 orthotopic tumor model) and displays no apparent detrimental effects in preliminary safety evaluation. Collectively, this study demonstrates the feasibility of exploiting DR6/APP interaction to regulate hematogenous tumor cells transendothelial migration and provides PEG-tAHP-DRI as a novel and promising inhibitor of DR6/APP interaction for developments of anti-hematogenous metastatic therapies.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Cell Communication/drug effects , Hematologic Neoplasms/drug therapy , Peptidomimetics/pharmacology , Receptors, Tumor Necrosis Factor/genetics , Amyloid beta-Protein Precursor/antagonists & inhibitors , Animals , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/pathology , Hematologic Neoplasms/blood , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Ligands , Melanoma, Experimental/drug therapy , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Neoplasm Metastasis , Peptidomimetics/chemistry , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Transendothelial and Transepithelial Migration/drug effects , Transendothelial and Transepithelial Migration/genetics
4.
J Am Chem Soc ; 143(2): 891-901, 2021 01 20.
Article in English | MEDLINE | ID: mdl-33398998

ABSTRACT

There is an urgent need for novel therapeutic approaches to treat Alzheimer's disease (AD) with the ability to both alleviate the clinical symptoms and halt the progression of the disease. AD is characterized by the accumulation of amyloid-ß (Aß) peptides which are generated through the sequential proteolytic cleavage of the amyloid precursor protein (APP). Previous studies reported that Mint2, a neuronal adaptor protein binding both APP and the γ-secretase complex, affects APP processing and formation of pathogenic Aß. However, there have been contradicting results concerning whether Mint2 has a facilitative or suppressive effect on Aß generation. Herein, we deciphered the APP-Mint2 protein-protein interaction (PPI) via extensive probing of both backbone H-bond and side-chain interactions. We also developed a proteolytically stable, high-affinity peptide targeting the APP-Mint2 interaction. We found that both an APP binding-deficient Mint2 variant and a cell-permeable PPI inhibitor significantly reduced Aß42 levels in a neuronal in vitro model of AD. Together, these findings demonstrate a facilitative role of Mint2 in Aß formation, and the combination of genetic and pharmacological approaches suggests that targeting Mint2 is a promising therapeutic strategy to reduce pathogenic Aß levels.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Protein Precursor/antagonists & inhibitors , Cadherins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Peptides/pharmacology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Cadherins/metabolism , Humans , Nerve Tissue Proteins/metabolism , Peptides/chemical synthesis , Peptides/chemistry , Protein Binding/drug effects
5.
Cancer Biother Radiopharm ; 36(9): 737-752, 2021 Nov.
Article in English | MEDLINE | ID: mdl-32343603

ABSTRACT

Background: Circular RNAs (circRNAs) have recently been reported to play essential roles in the progression of various cancers, including colorectal cancer (CRC). However, the roles of circRNA amyloid precursor-like protein 2 (circAPLP2) in CRC and its underlying mechanism have not been investigated. Materials and Methods: The expression levels of circAPLP2, microRNA-485-5p (miR-485-5p), and forkhead-box K1 (FOXK1) were determined by quantitative real-time polymerase chain reaction. Cell proliferation, apoptosis, migration, and invasion were assessed by methylthiazolyldiphenyl-tetrazolium bromide assay, flow cytometry, and transwell assay, respectively. Western blot assay was performed to measure the protein levels of proliferating cell nuclear antigen, Bcl-2, Bax, vimentin, E-cadherin, fibronectin, and FOXK1. The interaction between miR-485-5p and circAPLP2 or FOXK1 was predicted by starBase and verified by dual-luciferase reporter assay. A xenograft tumor model was established to confirm the functions of circAPLP2 in vivo. The lactate production was measured using lactate assay kit. Results: circAPLP2 expression was enhanced in CRC tissues and cells. circAPLP2 knockdown or miR-485-5p overexpression suppressed cell proliferation, migration, and invasion, whereas it promoted apoptosis in CRC cells, which was reversed by upregulating FOXK1. Moreover, miR-485-5p could directly bind to circAPLP2 and its downregulation reversed the suppressive effect of circAPLP2 knockdown on progression of CRC cells. In addition, FOXK1 was a downstream target of miR-485-5p. Furthermore, circAPLP2 modulated FOXK1 expression by sponging miR-485-5p in CRC cells. Besides, interference of circAPLP2 suppressed tumor growth in vivo and inhibited glycolysis in vitro by upregulating miR-485-5p and downregulating FOXK1. Conclusions: circAPLP2 knockdown inhibited CRC progression through regulating miR-485-5p/FOXK1 axis, providing a novel avenue for treatment of CRC.


Subject(s)
Amyloid beta-Protein Precursor , Colorectal Neoplasms , Forkhead Transcription Factors/metabolism , MicroRNAs/metabolism , Nerve Tissue Proteins , Amyloid beta-Protein Precursor/antagonists & inhibitors , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Down-Regulation , Drug Discovery , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques/methods , Humans , Male , Middle Aged , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Circular , Signal Transduction
6.
Exp Neurol ; 336: 113524, 2021 02.
Article in English | MEDLINE | ID: mdl-33159930

ABSTRACT

Ubiquitin (Ub) C-terminal hydrolase L1 (UCHL1) is a multifunctional protein that is expressed in neurons throughout brain at high levels. UCHL1 deletion is associated with axonal degeneration, progressive sensory motor ataxia, and premature death in mice. UCHL1 has been hypothesized to play a role in the pathogenesis of neurodegenerative diseases and recovery after neuronal injury. UCHL1 hydrolyzes Ub from polyubiquitinated (poly-Ub) proteins, but also may ligate Ub to select neuronal proteins, and interact with cytoskeletal proteins. These and other mechanisms have been hypothesized to underlie UCHL1's role in neurodegeneration and response to brain injury. A UCHL1 knockin mouse containing a C90A mutation (C90A) devoid of hydrolase activity was constructed. The C90A mouse did not develop the sensory and motor deficits, degeneration of the gracile nucleus and tract, or premature death as seen in UCHL1 deficient mice. C90A and wild type (WT) mice were subjected to the controlled cortical impact (CCI) model of traumatic brain injury (TBI), and cell death, axonal injury and behavioral outcome were assessed. C90A mice exhibited decreased spared tissue volume, greater loss of CA1 hippocampal neurons and greater axonal injury as detected using anti-amyloid precursor protein (APP) antibody and anti- non-phosphorylated neurofilament H (SMI-32) antibody immunohistochemistry after CCI compared to WT controls. Poly-Ub proteins and Beclin-1 were elevated after CCI in C90A mice compared to WT controls. Vestibular motor deficits assessed using the beam balance test resolved by day 5 after CCI in WT mice but not in C90A mice. These results suggest that the hydrolase activity of UCHL1 does not account for the progressive neurodegeneration and premature death seen in mice that do not express full length UCHL1. The hydrolase activity of UCHL1 contributes to the function of the ubiquitin proteasome pathway (UPP), ameliorates activation of autophagy, and improves motor recovery after CCI. Thus, UCHL1 hydrolase activity plays an important role in acute injury response after TBI.


Subject(s)
Axons/pathology , Brain Injuries, Traumatic/pathology , Cell Death/drug effects , Neurons/pathology , Ubiquitin Thiolesterase/genetics , Amyloid beta-Protein Precursor/antagonists & inhibitors , Animals , Autophagy , Beclin-1/metabolism , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/psychology , CA1 Region, Hippocampal/pathology , Cell Death/genetics , Gene Knock-In Techniques , Mice , Mutation/genetics , Psychomotor Performance , Signal Transduction/genetics , Ubiquitination
7.
Nat Neurosci ; 23(12): 1580-1588, 2020 12.
Article in English | MEDLINE | ID: mdl-33199898

ABSTRACT

Amyloid-ß (Aß) deposits are a relatively late consequence of Aß aggregation in Alzheimer's disease. When pathogenic Aß seeds begin to form, propagate and spread is not known, nor are they biochemically defined. We tested various antibodies for their ability to neutralize Aß seeds before Aß deposition becomes detectable in Aß precursor protein-transgenic mice. We also characterized the different antibody recognition profiles using immunoprecipitation of size-fractionated, native, mouse and human brain-derived Aß assemblies. At least one antibody, aducanumab, after acute administration at the pre-amyloid stage, led to a significant reduction of Aß deposition and downstream pathologies 6 months later. This demonstrates that therapeutically targetable pathogenic Aß seeds already exist during the lag phase of protein aggregation in the brain. Thus, the preclinical phase of Alzheimer's disease-currently defined as Aß deposition without clinical symptoms-may be a relatively late manifestation of a much earlier pathogenic seed formation and propagation that currently escapes detection in vivo.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/antagonists & inhibitors , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacology , Brain Chemistry , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Neurofilament Proteins/cerebrospinal fluid , Plaque, Amyloid/pathology , Tissue Extracts/pharmacology
8.
J Alzheimers Dis ; 77(1): 99-112, 2020.
Article in English | MEDLINE | ID: mdl-32804152

ABSTRACT

BACKGROUND: Transthyretin (TTR) is a tetrameric, amyloid-ß (Aß)-binding protein, which reduces Aß toxicity. The TTR/Aß interaction can be enhanced by a series of small molecules that stabilize its tetrameric form. Hence, TTR stabilizers might act as disease-modifying drugs in Alzheimer's disease. OBJECTIVE: We monitored the therapeutic efficacy of two TTR stabilizers, iododiflunisal (IDIF), which acts as small-molecule chaperone of the TTR/Aß interaction, and tolcapone, which does not behave as a small-molecule chaperone, in an animal model of Alzheimer's disease using positron emission tomography (PET). METHODS: Female mice (AßPPswe/PS1A246E/TTR+/-) were divided into 3 groups (n = 7 per group): IDIF-treated, tolcapone-treated, and non-treated. The oral treatment (100 mg/Kg/day) was started at 5 months of age. Treatment efficacy assessment was based on changes in longitudinal deposition of Aß in the hippocampus (HIP) and the cortex (CTX) and determined using PET-[18F]florbetaben. Immunohistochemical analysis was performed at age = 14 months. RESULTS: Standard uptake values relative to the cerebellum (SUVr) of [18F]florbetaben in CTX and HIP of non-treated animals progressively increased from age = 5 to 11 months and stabilized afterwards. In contrast, [18F]florbetaben uptake in HIP of IDIF-treated animals remained constant between ages = 5 and 11 months and significantly increased at 14 months. In the tolcapone-treated group, SUVr progressively increased with time, but at lower rate than in the non-treated group. No significant treatment effect was observed in CTX. Results from immunohistochemistry matched the in vivo data at age = 14 months. CONCLUSION: Our work provides encouraging preliminary results on the ability of small-molecule chaperones to ameliorate Aß deposition in certain brain regions.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/antagonists & inhibitors , Diflunisal/analogs & derivatives , Hippocampus/drug effects , Molecular Imaging/methods , Administration, Oral , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Diflunisal/administration & dosage , Female , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Longitudinal Studies , Mice , Mice, 129 Strain , Mice, Inbred C3H , Mice, Transgenic , Positron Emission Tomography Computed Tomography/methods
9.
Brain Res Bull ; 161: 197-212, 2020 08.
Article in English | MEDLINE | ID: mdl-32389800

ABSTRACT

Obesity and consumption of diet rich in fat are known to contribute to the development of Alzheimer's disease (AD) which is a complex and multifactorial neurodegenerative disease and a leading cause of mortality with unmet medical needs. Hypercholesterolemia was discovered to increase neuropathological changes along with cognitive decline in AD mouse models but still the underlying mechanism is elusive. Furthermore, isoprenoids, the crucial products of Mevalonate-pathway produced by the action of farnesyl pyrophosphate synthase (FPPS) enzyme, are also demonstrated to play a key role in AD. Nevertheless, bisphosphonates target this enzyme in order to treat osteoporosis and also found to alleviate dementia in such patients. As per the cited inhibitory action of alendronate, against acetylcholinesterase and cholesterol level, we hypothesized to explore the potential of alendronate against high fat diet (HFD) induced neuropathologies and cognitive disabilities in AD mouse model. Here we noticed that in mice provided with HFD for 14 weeks, spatial memory was compromised as interpreted in different behavioral paradigms. Together with cognitive depletion, there was observed a provoking effect on amyloid precursor protein (APP)-processing via amyloidogenic pathway due to enhanced ß-site APP cleaving enzyme-1 (BACE-1) level which in turn leads to augmented release of amyloid beta (Aß) in hippocampus of HFD mice. Relevant to these, significant elevation in hippocampal level of neuroinflammatory cytokines, oxidative stress markers and isoprenoids and serum cholesterol were also found after HFD exposure. Marked reversal of cognitive impairment, enhanced APP-processing, neuroinflammation along with other neuropathological alterations in hippocampus was demonstrated following oral administration of alendronate (1.76 mg/kg) for 15 days despite of HFD treatment. These changes were noted to be due to modulation of isoprenoids and cholesterol level by alendronate. Supporting these, histopathological analysis done by congo red revealed the reduced Aß deposition in hippocampus of drug treated HFD mice The current outcomes provide important implications for the contribution of Mevalonate-pathway and HFD for the onset of AD and also support alendronate as a prominent intervention for amelioration of AD-like pathologies.


Subject(s)
Alendronate/pharmacology , Amyloid beta-Protein Precursor/antagonists & inhibitors , Diet, High-Fat/adverse effects , Inflammation Mediators/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Alendronate/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Animals , Female , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Inflammation Mediators/metabolism , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Neuroprotective Agents/therapeutic use
10.
J Neurosci ; 40(27): 5161-5176, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32444385

ABSTRACT

Alterations of excitatory synaptic function are the strongest correlate to the pathologic disturbance of cognitive ability observed in the early stages of Alzheimer's disease (AD). This pathologic feature is driven by amyloid-ß oligomers (Aßos) and propagates from neuron to neuron. Here, we investigated the mechanism by which Aßos affect the function of synapses and how these alterations propagate to surrounding healthy neurons. We used complementary techniques ranging from electrophysiological recordings and molecular biology to confocal microscopy in primary cortical cultures, and from acute hippocampal and cortical slices from male wild-type and amyloid precursor protein (APP) knock-out (KO) mice to assess the effects of Aßos on glutamatergic transmission, synaptic plasticity, and dendritic spine structure. We showed that extracellular application of Aßos reduced glutamatergic synaptic transmission and long-term potentiation. These alterations were not observed in APP KO neurons, suggesting that APP expression is required. We demonstrated that Aßos/APP interaction increases the amyloidogenic processing of APP leading to intracellular accumulation of newly produced Aßos. Intracellular Aßos participate in synaptic dysfunctions as shown by pharmacological inhibition of APP processing or by intraneuronal infusion of an antibody raised against Aßos. Furthermore, we provide evidence that following APP processing, extracellular release of Aßos mediates the propagation of the synaptic pathology characterized by a decreased spine density of neighboring healthy neurons in an APP-dependent manner. Together, our data unveil a complementary role for Aßos in AD, while intracellular Aßos alter synaptic function, extracellular Aßos promote a vicious cycle that propagates synaptic pathology from diseased to healthy neurons.SIGNIFICANCE STATEMENT Here we provide the proof that a vicious cycle between extracellular and intracellular pools of Aß oligomers (Aßos) is required for the spreading of Alzheimer's disease (AD) pathology. We showed that extracellular Aßos propagate excitatory synaptic alterations by promoting amyloid precursor protein (APP) processing. Our results also suggest that subsequent to APP cleavage two pools of Aßos are produced. One pool accumulates inside the cytosol, inducing the loss of synaptic plasticity potential. The other pool is released into the extracellular space and contributes to the propagation of the pathology from diseased to healthy neurons. Pharmacological strategies targeting the proteolytic cleavage of APP disrupt the relationship between extracellular and intracellular Aß, providing a therapeutic approach for the disease.


Subject(s)
Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/metabolism , Neuronal Plasticity/drug effects , Neurons/metabolism , Synapses/drug effects , Amyloid beta-Protein Precursor/antagonists & inhibitors , Animals , Antibodies, Blocking/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Extracellular Space/drug effects , Extracellular Space/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Histidine/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Patch-Clamp Techniques , Primary Cell Culture , Synaptic Transmission/drug effects
11.
J Alzheimers Dis ; 74(3): 937-950, 2020.
Article in English | MEDLINE | ID: mdl-32116258

ABSTRACT

Alzheimer's disease (AD) is characterized by progressive cognitive decline and pathologically by the accumulation of amyloid-ß (Aß) and tau hyperphosphorylation causing neurodegeneration and neuroinflammation. Current AD treatments do not stop or reverse the disease progression, highlighting the need for more effective therapeutics. The phytocannabinoid cannabidiol (CBD) has demonstrated antioxidant, anti-inflammatory, and neuroprotective properties. Furthermore, chronic CBD treatment (20 mg/kg) reverses social and object recognition memory deficits in the AßPPxPS1 transgenic mouse model with only limited effects on AD-relevant brain pathology. Importantly, studies have indicated that CBD works in a dose-dependent manner. Thus, this study determined the chronic effects of 50 mg/kg CBD in male AßPPxPS1 mice. 12-month-old mice were treated with 50 mg/kg CBD or vehicle via daily intraperitoneal injections for 3 weeks prior to behavioral testing. A variety of cognitive domains including object and social recognition, spatial and fear-associated memory were evaluated. Pathological brain analyses for AD-relevant markers were conducted using ELISA and western blot. Vehicle-treated male AßPPxPS1 mice demonstrated impaired social recognition memory and reversal spatial learning. These deficits were restored after CBD treatment. Chronic CBD tended to reduce insoluble Aß40 levels in the hippocampus of AßPPxPS1 mice but had no effect on neuroinflammation, neurodegeneration, or PPARγ markers in the cortex. This study demonstrates that therapeutic-like effects of 50 mg/kg CBD on social recognition memory and spatial learning deficits in AßPPxPS1 mice are accompanied by moderate brain region-specific reductions in insoluble Aß40 levels. The findings emphasize the clinical relevance of CBD treatment in AD; however, the underlying mechanisms involved require further investigation.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Cannabidiol/therapeutic use , Cognition/drug effects , Peptide Fragments/metabolism , Presenilin-1/genetics , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/antagonists & inhibitors , Animals , Brain/pathology , Dose-Response Relationship, Drug , Fear/drug effects , Fear/psychology , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1/antagonists & inhibitors , Recognition, Psychology , Social Behavior , Space Perception/drug effects
12.
Curr Med Sci ; 40(6): 999-1008, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33428127

ABSTRACT

Alzheimer's disease (AD) is a chronic neurodegenerative disease that mainly causes dementia. It is a serious threat to the health of the global elderly population. Considerable money and effort has been invested in the development of drug therapy for AD worldwide. Many drug therapies are currently under development or in clinical trials, based on two known mechanisms of AD, namely, Aß toxicity and the abnormal Tau hyperphosphorylation. Numerous drugs are also being developed for other AD associated mechanisms such as neuroinflammation, neurotransmitter imbalance, oxidative damage and mitochondrial dysfunction, neuron loss and degeneration. Even so, the number of drugs that can successfully improve symptoms or delay the progression of the disease remains very limited. However, multi-drug combinations may provide a new avenue for drug therapy for AD. In addition, early diagnosis of AD and timely initiation of treatment may allow drugs that act on the early pathological processes of AD to help improve the symptoms and prevent the progression of the condition.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/metabolism , tau Proteins/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/antagonists & inhibitors , Clinical Trials as Topic , Disease Progression , Humans , Molecular Targeted Therapy , Phosphorylation/drug effects , Signal Transduction/drug effects , tau Proteins/antagonists & inhibitors
13.
J Alzheimers Dis ; 72(3): 761-771, 2019.
Article in English | MEDLINE | ID: mdl-31640096

ABSTRACT

Alzheimer's disease (AD) is a devastating neurodegenerative disease with no cure currently available. A pathological hallmark of AD is accumulation and deposition of amyloid-ß protein (Aß), a ∼4 kDa peptide generated through serial cleavage of the amyloid-ß protein precursor (AßPP) by ß- and γ-secretases. Curcumin is a natural compound primarily found in the widely used culinary spice, turmeric, which displays therapeutic potential for AD. Recently, we reported the development of curcumin analogs and identified a lead compound, curcumin-like compound-R17 (CLC-R17), that significantly attenuates Aß deposition in an AD transgenic mouse model. Here, we elucidated the mechanisms of this analog on Aß levels and AßPP processing using cell models of AD. Using biochemical methods and our recently developed nanoplasmonic fiber tip probe technology, we showed that the lead compound potently lowers Aß levels in conditioned media and reduces oligomeric amyloid levels in the cells. Furthermore, like curcumin, the lead compound attenuates the maturation of AßPP in the secretory pathway. Interestingly, it upregulated α-secretase processing of AßPP and inhibited ß-secretase processing of AßPP by decreasing BACE1 protein levels. Collectively, our data reveal mechanisms of a promising curcumin analog in reducing Aß levels, which strongly support its development as a potential therapeutic for AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Autophagy/drug effects , Curcumin/analogs & derivatives , Curcumin/pharmacology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Protein Precursor/antagonists & inhibitors , Animals , Autophagy/physiology , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Humans
14.
Free Radic Biol Med ; 145: 20-32, 2019 12.
Article in English | MEDLINE | ID: mdl-31536772

ABSTRACT

Alzheimer's disease (AD) is a common neurodegenerative disease that progressively impairs memory and cognition. Deposition of amyloid-ß (Aß) peptides is the most important pathophysiological hallmark of AD. Oxidative stress induced by generation of reactive oxygen species (ROS) is a prominent phenomenon in AD and known to occur early in the course of AD. Several reports suggest a relationship between change in redox status and AD pathology including progressive Aß deposition, glial cell activation, and inflammation. Galantamine is an acetylcholinesterase inhibitor and has been reported to have an oxidative stress inhibitory function. In the present study, galantamine was administered orally to AD model mice from before the appearance of Aß plaques (preplaque phase), and in vivo change in redox status of the brain was measured using electron paramagnetic resonance (EPR) imaging. Administration of galantamine from the preplaque phase ameliorated memory decline in Morris water maze test and novel object recognition test. Monitoring of the redox status of the brain using EPR imaging showed that galantamine treatment improved the unbalanced redox state. Additionally, galantamine administration enhanced microglial function to promote Aß clearance, reducing the Aß-positive area in the cortex and amount of insoluble Aß in the brain. In contrast, galantamine treatment from the preplaque phase suppressed the production of proinflammatory cytokines through neurotoxic microglial activity. Therefore, galantamine administration from the preplaque phase may have the potential of clinical application for the prevention of AD. In addition, our results demonstrate the usefulness of EPR imaging for speedy and quantitative evaluation of the efficacy of disease-modifying drugs for AD.


Subject(s)
Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/pharmacology , Galantamine/pharmacology , Oxidative Stress/drug effects , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/antagonists & inhibitors , Amyloid beta-Protein Precursor/genetics , Animals , Cognitive Behavioral Therapy , Disease Models, Animal , Electron Spin Resonance Spectroscopy , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/pathology , Mice , Microglia/drug effects , Microglia/pathology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/prevention & control , RNA-Binding Proteins/genetics , Reactive Oxygen Species/metabolism , Ribosomal Proteins/genetics
16.
Molecules ; 24(9)2019 May 06.
Article in English | MEDLINE | ID: mdl-31064071

ABSTRACT

As a longstanding problem, Alzheimer's disease (AD) has stymied researchers in the medical field with its increasing incidence and enormous treatment difficulty. Silymarin has always been valued by researchers for its good efficacy and safety in treating liver disease. Recent studies have shown that silymarin also has good pharmacological activity in the nervous system, especially for the treatment of AD. Silymarin can control the production of Aß by inhibiting the precursor substance of Aß (ß-amyloid precursor protein), and it can inhibit the polymerization of Aß. Silymarin can also increase the acetylcholine content in the nervous system by inhibiting cholinesterase activity. At the same time, it also has the effect of resisting oxidative stress and the inflammatory response of the nervous system. These pharmacological activities contribute to the inhibition of the onset of AD. The good efficacy of silymarin on AD and its high safety and availability give it huge potential for the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/antagonists & inhibitors , Silymarin/therapeutic use , Acetylcholine/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cholinesterase Inhibitors/pharmacology , Cholinesterases/metabolism , Humans , Molecular Structure , Nervous System/drug effects , Oxidative Stress/drug effects , Silymarin/pharmacology
17.
N Engl J Med ; 380(15): 1408-1420, 2019 Apr 11.
Article in English | MEDLINE | ID: mdl-30970186

ABSTRACT

BACKGROUND: Prodromal Alzheimer's disease offers an opportunity to test the effect of drugs that modify the deposition of amyloid in the brain before the onset of dementia. Verubecestat is an orally administered ß-site amyloid precursor protein-cleaving enzyme 1 (BACE-1) inhibitor that blocks production of amyloid-beta (Aß). The drug did not prevent clinical progression in a trial involving patients with mild-to-moderate dementia due to Alzheimer's disease. METHODS: We conducted a randomized, double-blind, placebo-controlled, 104-week trial to evaluate verubecestat at doses of 12 mg and 40 mg per day, as compared with placebo, in patients who had memory impairment and elevated brain amyloid levels but whose condition did not meet the case definition of dementia. The primary outcome was the change from baseline to week 104 in the score on the Clinical Dementia Rating Scale-Sum of Boxes (CDR-SB; scores range from 0 to 18, with higher scores indicating worse cognition and daily function). Secondary outcomes included other assessments of cognition and daily function. RESULTS: The trial was terminated for futility after 1454 patients had been enrolled; 485 had been assigned to receive verubecestat at a dose of 12 mg per day (the 12-mg group), 484 to receive verubecestat at a dose of 40 mg per day (the 40-mg group), and 485 to receive placebo. A total of 234 patients, 231 patients, and 239 patients per group, respectively, completed 104 weeks of the trial regimen. The estimated mean change from baseline to week 104 in the CDR-SB score was 1.65 in the 12-mg group, 2.02 in the 40-mg group, and 1.58 in the placebo group (P = 0.67 for the comparison between the 12-mg group and the placebo group and P = 0.01 for the comparison between the 40-mg group and the placebo group), suggesting a worse outcome in the higher-dose group than in the placebo group. The estimated rate of progression to dementia due to Alzheimer's disease was 24.5, 25.5, and 19.3 events per 100 patient-years in the 12-mg group, the 40-mg group, and the placebo group, respectively (hazard ratio for 40 mg vs. placebo, 1.38; 97.51% confidence interval, 1.07 to 1.79, not adjusted for multiple comparisons), favoring placebo. Adverse events were more common in the verubecestat groups than in the placebo group. CONCLUSIONS: Verubecestat did not improve clinical ratings of dementia among patients with prodromal Alzheimer's disease, and some measures suggested that cognition and daily function were worse among patients who received verubecestat than among those who received placebo. (Funded by Merck Sharp & Dohme; ClinicalTrials.gov number, NCT01953601.).


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Protein Precursor/antagonists & inhibitors , Cognitive Dysfunction/drug therapy , Cyclic S-Oxides/therapeutic use , Thiadiazines/therapeutic use , Aged , Amyloid beta-Peptides/analysis , Brain Chemistry , Cognitive Dysfunction/pathology , Cyclic S-Oxides/adverse effects , Disease Progression , Double-Blind Method , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/therapeutic use , Female , Hippocampus/pathology , Humans , Intention to Treat Analysis , Magnetic Resonance Imaging , Male , Organ Size , Plaque, Amyloid/diagnostic imaging , Positron-Emission Tomography , Prodromal Symptoms , Thiadiazines/adverse effects , Treatment Failure
18.
Neurochem Int ; 126: 195-202, 2019 06.
Article in English | MEDLINE | ID: mdl-30905744

ABSTRACT

Magnesium (Mg) is a crucial divalent cation with more than 300 cellular functions. This ion shows therapeutic properties in several neurological diseases. Although there are numerous basic evidences showing that Mg can inhibit pathological processes involved in neuroglial degeneration, this low-cost option is not well-considered in clinical research and practice for now. Nevertheless, none of the expensive drugs currently recommended by the classic guidelines (in addition to physiological rehabilitation) had shown exceptional effectiveness. Herein, focusing on Alzheimer's disease (AD), we analyze the therapeutic pathways that support the use of Mg for neurogenesis and neuroprotection. According to experimental findings reviewed, Mg shows interesting abilities to facilitate toxin clearance, reduce neuroinflammation, inhibit the pathologic processing of amyloid protein precursor (APP) as well as the abnormal tau protein phosphorylation, and to reverse the deregulation of N-methyl-D-aspartate receptors. Currently, some crucial details of the mechanisms involved in these proved effects remain elusive and clinical background is poor. Therefore, further studies are required to enable a better overview on pharmacodynamic targets of Mg and thus, to find optimal pharmacologic strategies for clinical use of this ion.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Magnesium/metabolism , tau Proteins/metabolism , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Protein Precursor/antagonists & inhibitors , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Humans , Magnesium/administration & dosage , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , tau Proteins/antagonists & inhibitors
19.
Mar Drugs ; 17(2)2019 Feb 18.
Article in English | MEDLINE | ID: mdl-30781690

ABSTRACT

Astaxanthin (AXT), a xanthophyll carotenoid compound, has potent antioxidant, anti-inflammatory and neuroprotective properties. Neuroinflammation and oxidative stress are significant in the pathogenesis and development of Alzheimer's disease (AD). Here, we studied whether AXT could alleviate neuroinflammation, oxidative stress and memory loss in lipopolysaccharide (LPS) administered mice model. Additionally, we investigated the anti-oxidant activity and the anti-neuroinflammatory response of AXT in LPS-treated BV-2 microglial cells. The AXT administration ameliorated LPS-induced memory loss. This effect was associated with the reduction of LPS-induced expression of inflammatory proteins, as well as the production of reactive oxygen species (ROS), nitric oxide (NO), cytokines and chemokines both in vivo and in vitro. AXT also reduced LPS-induced ß-secretase and Aß1⁻42 generation through the down-regulation of amyloidogenic proteins both in vivo and in vitro. Furthermore, AXT suppressed the DNA binding activities of the signal transducer and activator of transcription 3 (STAT3). We found that AXT directly bound to the DNA- binding domain (DBD) and linker domain (LD) domains of STAT3 using docking studies. The oxidative stress and inflammatory responses were not downregulated in BV-2 cells transfected with DBD-null STAT3 and LD-null STAT3. These results indicated AXT inhibits LPS-induced oxidant activity, neuroinflammatory response and amyloidogenesis via the blocking of STAT3 activity through direct binding.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Inflammation/chemically induced , Inflammation/prevention & control , Lipopolysaccharides , Memory Disorders/prevention & control , STAT3 Transcription Factor/drug effects , Amyloid beta-Protein Precursor/antagonists & inhibitors , Amyloid beta-Protein Precursor/biosynthesis , Animals , Antioxidants/pharmacology , Avoidance Learning/drug effects , Cell Line , Maze Learning/drug effects , Memory Disorders/chemically induced , Mental Recall/drug effects , Mice , Mice, Inbred ICR , Microglia/drug effects , Oxidative Stress/drug effects , Signal Transduction/drug effects , Xanthophylls/therapeutic use
20.
J Alzheimers Dis ; 68(1): 115-126, 2019.
Article in English | MEDLINE | ID: mdl-30689563

ABSTRACT

The degeneration in the locus coeruleus associated with Alzheimer's disease suggests an involvement of the noradrenergic system in the disease pathogenesis. The role of depleted norepinephrine was tested in adult and aged rhesus macaques to develop a potential model for testing Alzheimer's disease interventions. Monkeys were injected with the noradrenergic neurotoxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP4) or vehicle at 0, 3, and 6 months; brains were harvested at 9 months. Reduced norepinephrine in the locus coeruleus was accompanied by decreased dopamine ß-hydroxylase staining and increased amyloid-ß load in the aged group, and the proportion of potentially toxic amyloid-ß42 peptide was increased. Immunohistochemistry revealed no effects on microglia or astrocytes. DSP4 treatment altered amyloid processing, but these changes were not associated with the induction of chronic neuroinflammation. These findings suggest norepinephrine deregulation is an essential component of a nonhuman primate model of Alzheimer's disease, but further refinement is necessary.


Subject(s)
Amyloid beta-Peptides/metabolism , Benzylamines/pharmacology , Locus Coeruleus/metabolism , Neurotransmitter Uptake Inhibitors/pharmacology , Norepinephrine/metabolism , Peptide Fragments/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Protein Precursor/antagonists & inhibitors , Amyloid beta-Protein Precursor/metabolism , Animals , Female , Locus Coeruleus/drug effects , Macaca mulatta , Norepinephrine/antagonists & inhibitors , Peptide Fragments/antagonists & inhibitors , Random Allocation
SELECTION OF CITATIONS
SEARCH DETAIL
...