Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 205
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Article in English | MEDLINE | ID: mdl-33431651

ABSTRACT

Alzheimer's disease (AD), the most common cause of dementia and neurodegeneration in the elderly, is characterized by deterioration of memory and executive and motor functions. Neuropathologic hallmarks of AD include neurofibrillary tangles (NFTs), paired helical filaments, and amyloid plaques. Mutations in the microtubule-associated protein Tau, a major component of the NFTs, cause its hyperphosphorylation in AD. We have shown that signaling by the gaseous molecule hydrogen sulfide (H2S) is dysregulated during aging. H2S signals via a posttranslational modification termed sulfhydration/persulfidation, which participates in diverse cellular processes. Here we show that cystathionine γ-lyase (CSE), the biosynthetic enzyme for H2S, binds wild type Tau, which enhances its catalytic activity. By contrast, CSE fails to bind Tau P301L, a mutant that is present in the 3xTg-AD mouse model of AD. We further show that CSE is depleted in 3xTg-AD mice as well as in human AD brains, and that H2S prevents hyperphosphorylation of Tau by sulfhydrating its kinase, glycogen synthase kinase 3ß (GSK3ß). Finally, we demonstrate that sulfhydration is diminished in AD, while administering the H2S donor sodium GYY4137 (NaGYY) to 3xTg-AD mice ameliorates motor and cognitive deficits in AD.


Subject(s)
Alzheimer Disease/drug therapy , Cystathionine gamma-Lyase/genetics , Glycogen Synthase Kinase 3 beta/genetics , Hydrogen Sulfide/pharmacology , Morpholines/pharmacology , Neuroprotective Agents/pharmacology , Organothiophosphorus Compounds/pharmacology , tau Proteins/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Cystathionine gamma-Lyase/metabolism , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/metabolism , HEK293 Cells , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice , Mice, Transgenic , Mutation , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Phosphorylation , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/prevention & control , Protein Binding , Protein Processing, Post-Translational , Sulfates/metabolism , tau Proteins/metabolism
2.
Cell Mol Neurobiol ; 41(3): 449-458, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32385548

ABSTRACT

Glaucoma is a neurodegenerative disorder that leads to the slow degeneration of retinal ganglion cells, and results in damage to the optic nerve and concomitant vision loss. As in other disorders affecting the viability of central nervous system neurons, neurons affected by glaucoma do not have the ability to regenerate after injury. Recent studies indicate a critical role for optic nerve head astrocytes (ONHAs) in this process of retinal ganglion cell degeneration. Cleavage of tau, a microtubule stabilizing protein and constituent of neurofibrillary tangles (NFT), plays a major part in the mechanisms that lead to toxicity in CNS neurons and astrocytes. Here, we tested the hypothesis that estrogen, a pleiotropic neuro- and cytoprotectant with high efficacy in the CNS, prevents tau cleavage, and hence, protects ONHAs against cell damage caused by oxidative stress. Our results indicate that estrogen prevents caspase-3 mediated tau cleavage, and thereby decreases the levels of the resulting form of proteolytically cleaved tau protein, which leads to a decrease in NFT formation, which requires proteolytically cleaved tau protein. Overall, our data propose that by stopping the reduction of estrogen levels involved with aging the sensitivity of the optic nerve to glaucomatous damage might be reduced. Furthermore, our data suggest that therapeutic use of estrogen may be beneficial in slowing or preventing the onset or severity of neurodegenerative diseases such as glaucoma and potentially also other degenerative diseases of the CNS through direct control of posttranslational modifications of tau protein.


Subject(s)
Astrocytes/pathology , Caspase 3/metabolism , Estrogens/pharmacology , Optic Disk/pathology , Oxidative Stress , Protective Agents/pharmacology , Protein Aggregates , tau Proteins/metabolism , Animals , Astrocytes/drug effects , Astrocytes/enzymology , Biomarkers/metabolism , Cell Death/drug effects , Glial Fibrillary Acidic Protein/metabolism , Male , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/metabolism , Oxidative Stress/drug effects , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Aggregates/drug effects , Rats, Inbred BN , tert-Butylhydroperoxide/pharmacology
3.
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
4.
Brain ; 143(1): 359-373, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31782760

ABSTRACT

Failure of Alzheimer's disease clinical trials to improve or stabilize cognition has led to the need for a better understanding of the driving forces behind cognitive decline in the presence of active disease processes. To dissect contributions of individual pathologies to cognitive function, we used the TgF344-AD rat model, which recapitulates the salient hallmarks of Alzheimer's disease pathology observed in patient populations (amyloid, tau inclusions, frank neuronal loss, and cognitive deficits). scyllo-Inositol treatment attenuated amyloid-ß peptide in disease-bearing TgF344-AD rats, which rescued pattern separation in the novel object recognition task and executive function in the reversal learning phase of the Barnes maze. Interestingly, neither activities of daily living in the burrowing task nor spatial memory in the Barnes maze were rescued by attenuating amyloid-ß peptide. To understand the pathological correlates leading to behavioural rescue, we examined the neuropathology and in vivo electrophysiological signature of the hippocampus. Amyloid-ß peptide attenuation reduced hippocampal tau pathology and rescued adult hippocampal neurogenesis and neuronal function, via improvements in cross-frequency coupling between theta and gamma bands. To investigate mechanisms underlying the persistence of spatial memory deficits, we next examined neuropathology in the entorhinal cortex, a region whose input to the hippocampus is required for spatial memory. Reduction of amyloid-ß peptide in the entorhinal cortex had no effect on entorhinal tau pathology or entorhinal-hippocampal neuronal network dysfunction, as measured by an impairment in hippocampal response to entorhinal stimulation. Thus, rescue or not of cognitive function is dependent on regional differences of amyloid-ß, tau and neuronal network dysfunction, demonstrating the importance of staging disease in patients prior to enrolment in clinical trials. These results further emphasize the need for combination therapeutic approaches across disease progression.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/drug effects , Cognition/drug effects , Entorhinal Cortex/drug effects , Hippocampus/drug effects , Inositol/pharmacology , Spatial Memory/drug effects , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Entorhinal Cortex/metabolism , Entorhinal Cortex/pathology , Executive Function/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Maze Learning , Neural Pathways , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/pathology , Neurogenesis/drug effects , Rats , Rats, Transgenic , Recognition, Psychology/drug effects , Reversal Learning/drug effects
5.
Bioorg Chem ; 113: 105022, 2021 08.
Article in English | MEDLINE | ID: mdl-34098397

ABSTRACT

In this study, polyhydroxyisoflavones that directly prevent the aggregation of both amyloid ß (Aß) and tau were expediently synthesized via divergent Pd(0)-catalyzed Suzuki-Miyaura coupling and then biologically evaluated. By preliminary structure-activity relationship studies using thioflavin T (ThT) assays, an ortho-catechol containing isoflavone scaffold was proven to be crucial for preventing both Aß aggregation and tau-mediated neurofibrillary tangle formation. Additional TEM experiment confirmed that ortho-catechol containing isoflavone 4d significantly prevented the aggregation of both Aß and tau. To investigate the mode of action (MOA) of 4d, which possesses an ortho-catechol moiety, 1H-15N HSQC NMR analysis was thoroughly performed and the result indicated that 4d could directly inhibit both the formation of Aß42 fibrils and the formation of tau-derived neurofibrils, probably through the catechol-mediated nucleation of tau. Finally, 4d was demonstrated to alleviate cognitive impairment and pathologies related to Alzheimer's disease in a 5XFAD transgenic mouse model.


Subject(s)
Catechols/chemistry , Isoflavones/chemistry , Neuroprotective Agents/chemistry , tau Proteins/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Drug Design , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/metabolism , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Protein Aggregates/drug effects , tau Proteins/antagonists & inhibitors
6.
Int J Mol Sci ; 22(21)2021 Nov 08.
Article in English | MEDLINE | ID: mdl-34769495

ABSTRACT

The neuropathological hallmarks of Alzheimer's disease (AD) are senile plaques (SPs), which are composed of amyloid ß protein (Aß), and neurofibrillary tangles (NFTs), which consist of highly phosphorylated tau protein. As bio-metal imbalance may be involved in the formation of NFT and SPs, metal regulation may be a direction for AD treatment. Clioquinol (CQ) is a metal-protein attenuating compound with mild chelating effects for Zn2+ and Cu2+, and CQ can not only detach metals from SPs, but also decrease amyloid aggregation in the brain. Previous studies suggested that Cu2+ induces the hyperphosphorylation of tau. However, the effects of CQ on tau were not fully explored. To examine the effects of CQ on tau metabolism, we used a human neuroblastoma cell line, M1C cells, which express wild-type tau protein (4R0N) via tetracycline-off (TetOff) induction. In a morphological study and ATP assay, up to 10 µM CQ had no effect on cell viability; however, 100 µM CQ had cytotoxic effects. CQ decreased accumulation of Cu+ in the M1C cells (39.4% of the control), and both total and phosphorylated tau protein. It also decreased the activity of c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38 MAPK) (37.3% and 60.7% levels of the control, respectively), which are tau kinases. Of note, activation of protein phosphatase 2A (PP2A), which is a tau phosphatase, was also observed after CQ treatment. Fractionation experiments demonstrated a reduction of oligomeric tau in the tris insoluble, sarkosyl soluble fraction by CQ treatment. CQ also decreased caspase-cleaved tau, which accelerated the aggregation of tau protein. CQ activated autophagy and proteasome pathways, which are considered important for the degradation of tau protein. Although further studies are needed to elucidate the mechanisms responsible for the effects of CQ on tau, CQ may shed light on possible AD therapeutics.


Subject(s)
Alzheimer Disease/drug therapy , Clioquinol/pharmacology , Gene Expression Regulation/drug effects , Neurofibrillary Tangles/drug effects , Protein Multimerization , tau Proteins/chemistry , tau Proteins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Autophagy , Cell Line, Tumor , Copper/chemistry , Humans , Neurofibrillary Tangles/metabolism , Phosphorylation , Protein Phosphatase 2/metabolism
7.
Int J Mol Sci ; 21(13)2020 Jun 29.
Article in English | MEDLINE | ID: mdl-32610577

ABSTRACT

Aging is an ineluctable law of life. During the process of aging, the occurrence of neurodegenerative disorders is prevalent in the elderly population and the predominant type of dementia is Alzheimer's disease (AD). The clinical symptoms of AD include progressive memory loss and impairment of cognitive functions that interfere with daily life activities. The predominant neuropathological features in AD are extracellular ß-amyloid (Aß) plaque deposition and intracellular neurofibrillary tangles (NFTs) of hyperphosphorylated Tau. Because of its complex pathobiology, some tangible treatment can only ameliorate the symptoms, but not prevent the disease altogether. Numerous drugs during pre-clinical or clinical studies have shown no positive effect on the disease outcome. Therefore, understanding the basic pathophysiological mechanism of AD is imperative for the rational design of drugs that can be used to prevent this disease. Drosophila melanogaster has emerged as a highly efficient model system to explore the pathogenesis and treatment of AD. In this review we have summarized recent advancements in the pharmacological research on AD using Drosophila as a model species, discussed feasible treatment strategies and provided further reference for the mechanistic study and treatment of age-related AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Aging/physiology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/physiology , Animals , Disease Models, Animal , Drosophila melanogaster/metabolism , Humans , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/metabolism , Pharmacological Phenomena/drug effects , Pharmacological Phenomena/physiology , Plaque, Amyloid/pathology , tau Proteins/metabolism
8.
J Biol Chem ; 293(8): 2888-2902, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29282295

ABSTRACT

Amyloid-ß (Aß) and human islet amyloid polypeptide (hIAPP) aggregate to form amyloid fibrils that deposit in tissues and are associated with Alzheimer's disease (AD) and type II diabetes (T2D), respectively. Individuals with T2D have an increased risk of developing AD, and conversely, AD patients have an increased risk of developing T2D. Evidence suggests that this link between AD and T2D might originate from a structural similarity between aggregates of Aß and hIAPP. Using the cryoEM method microelectron diffraction, we determined the atomic structures of 11-residue segments from both Aß and hIAPP, termed Aß(24-34) WT and hIAPP(19-29) S20G, with 64% sequence similarity. We observed a high degree of structural similarity between their backbone atoms (0.96-Å root mean square deviation). Moreover, fibrils of these segments induced amyloid formation through self- and cross-seeding. Furthermore, inhibitors designed for one segment showed cross-efficacy for full-length Aß and hIAPP and reduced cytotoxicity of both proteins, although by apparently blocking different cytotoxic mechanisms. The similarity of the atomic structures of Aß(24-34) WT and hIAPP(19-29) S20G offers a molecular model for cross-seeding between Aß and hIAPP.


Subject(s)
Amyloid beta-Peptides/metabolism , Islet Amyloid Polypeptide/metabolism , Models, Molecular , Neurofibrillary Tangles/metabolism , Peptide Fragments/metabolism , Amino Acid Substitution , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Animals , Cell Line, Tumor , Computational Biology , Crystallography, X-Ray , Drug Design , HEK293 Cells , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/ultrastructure , Islet Amyloid Polypeptide/antagonists & inhibitors , Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/genetics , Mice , Microscopy, Electron, Transmission , Mutation , Neurofibrillary Tangles/chemistry , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/ultrastructure , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neurons/ultrastructure , Nootropic Agents/chemistry , Nootropic Agents/pharmacology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Protein Aggregation, Pathological/prevention & control , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure
9.
Neurobiol Dis ; 130: 104519, 2019 10.
Article in English | MEDLINE | ID: mdl-31233882

ABSTRACT

The intraneuronal aggregates of hyperphosphorylated and misfolded tau (neurofibrillary tangles, NFTs) cause a stereotypical spatiotemporal Alzheimer's disease (AD) progression that correlates with the severity of the associated cognitive decline. Kinase activity contributes to the balance between neuron survival and cell death. Hyperactivation of kinases including the conventional protein kinase C (PKC) is a defective molecular event accompanying associative memory loss, tau phosphorylation, and progression of AD or related neurodegenerative diseases. Here, we investigated the ability of small therapeutic compounds (a custom library) to improve tau-induced rough-eye phenotype in a Drosophila melanogaster model of frontotemporal dementia. We also assessed the tau phosphorylation in vivo and selected hit compounds. Among the potential hits, we investigated Ro 31-8220, described earlier as a potent PKCα inhibitor. Ro 31-8220 robustly improved the rough-eye phenotype, reduced phosphorylated tau species in vitro and in vivo, reversed tau-induced memory impairment, and improved the fly motor functions. In a human neuroblastoma cell line, Ro 31-8220 reduced the PKC activity and the tau phosphorylation pattern, but we also have to acknowledge the compound's wide range of biological activity. Nevertheless, Ro 31-8220 is a novel therapeutic mitigator of tau-induced neurotoxocity.


Subject(s)
Frontotemporal Dementia/metabolism , Indoles/pharmacology , Neurofibrillary Tangles/drug effects , Neurons/drug effects , tau Proteins/metabolism , Animals , Disease Models, Animal , Drosophila melanogaster , Drug Evaluation, Preclinical , Neurofibrillary Tangles/metabolism , Neurons/metabolism , Phosphorylation/drug effects
10.
Annu Rev Med ; 68: 413-430, 2017 01 14.
Article in English | MEDLINE | ID: mdl-28099083

ABSTRACT

Alzheimer's disease (AD) is the primary cause of age-related dementia. Effective strategies to prevent and treat AD remain elusive despite major efforts to understand its basic biology and clinical pathophysiology. Significant investments in therapeutic drug discovery programs over the past two decades have yielded some important insights but no blockbuster drugs to alter the course of disease. Because significant memory loss and cognitive decline are associated with neuron death and loss of gray matter, especially in the frontal cortex and hippocampus, some focus in drug development has shifted to early prevention of cellular pathology. Although clinical trial design is challenging, due in part to a lack of robust biomarkers with predictive value, some optimism has come from the identification and study of inherited forms of early-onset AD and genetic risk factors that provide insights about molecular pathophysiology and potential drug targets. In addition, better understanding of the Aß amyloid pathway and the tau pathway-leading to amyloid plaques and neurofibrillary tangles, respectively, which are histopathological hallmarks of AD-continues to drive significant drug research and development programs. The main focus of this review is to summarize the most recent basic biology, biochemistry, and pharmacology that serve as a foundation for more than 50 active advanced-phase clinical trials for AD prevention and therapy.


Subject(s)
Aging/physiology , Alzheimer Disease/physiopathology , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Alzheimer Disease/drug therapy , Animals , Cholinesterase Inhibitors/therapeutic use , Excitatory Amino Acid Antagonists/therapeutic use , Humans , Immunotherapy , Memantine/therapeutic use , Metabolic Diseases , Microglia/physiology , Molecular Chaperones/metabolism , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/metabolism , Neurogenic Inflammation/drug therapy , Neuroimmunomodulation , tau Proteins/metabolism
11.
Biochem Biophys Res Commun ; 495(1): 1034-1040, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29175324

ABSTRACT

Type 2 diabetes mellitus (T2DM) has been identified as a high risk factor for Alzheimer's disease (AD). The impairment of insulin signaling has been found in AD brain. Glucagon-like peptide-1 (GLP-1) is an incretin hormone, normalises insulin signaling and acts as a neuroprotective growth factor. We have previously shown that the long-lasting GLP-1 receptor (GLP-1R) agonist lixisenatide plays an important role in memory formation, synaptic plasticity and cell proliferation of rats. In the follow-up study, we analysed the neuroprotective effect and mechanism of lixisenatide, injected for 60 days at 10 nmol/kg i.p. once daily in APP/PS1/tau female mice and C57BL/6J female mice (as control) aged 12 month. The results showed that lixisenatide could reduce amyloid plaques, neurofibrillary tangles and neuroinflammation in the hippocampi of 12-month-old APP/PS1/tau female mice; activation of PKA-CREB signaling pathway and inhibition of p38-MAPK might be the important mechanisms in the neuroprotective function of lixisenatide. The study demonstrated that GLP-1R agonists such as lixisenatide might have the potential to be developed as a novel therapy for AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Encephalitis/drug therapy , Encephalitis/metabolism , Neurofibrillary Tangles/drug effects , Peptides/administration & dosage , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Dose-Response Relationship, Drug , Encephalitis/pathology , Female , Glucagon-Like Peptide-1 Receptor/agonists , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neuroprotective Agents/administration & dosage , Treatment Outcome
12.
Biochim Biophys Acta Mol Basis Dis ; 1864(6 Pt A): 2143-2153, 2018 06.
Article in English | MEDLINE | ID: mdl-29604335

ABSTRACT

Soluble form of the prion protein (PrP) has been previously shown to interact with amyloid-ß (Aß) peptides, suppressing their fibrillization as well as toxicity, which indicates that this protein may play a protective role in Alzheimer's disease (AD). The shortest known PrP fragment retaining all of these properties corresponds to physiologically generated proteolytic polypeptide PrP23-110/111, called N1. Here we have identified two N1-derived synthetic peptides, encompassing residues 23-50 (PrP23-50) and 90-112 (PrP90-112), which bind to Aß1-42 protofibrillar oligomers as well as amyloid fibrils. We found that, akin to N1, the abovementioned synthetic peptides not only reduce the initial rate of Aß fibrillization, but also alter the aggregation pathway of Aß, inhibiting formation of protofibrillar oligomers and facilitating amorphous aggregation. Furthermore, our data show that N1, PrP23-50 and PrP90-112 protect cultured hippocampal neurons from neurotoxic effects of Aß oligomers, preventing oligomers-induced retraction of neurites and loss of cell membrane integrity. The above PrP fragments can also attenuate neuronal intake of Aß. Our results strongly suggest that synthetic peptides such as PrP23-50 and PrP90-112 can be useful in designing a novel class of therapeutics in AD.


Subject(s)
Alzheimer Disease/drug therapy , Neurofibrillary Tangles/drug effects , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Protein Aggregation, Pathological/drug therapy , Alzheimer Disease/pathology , Amyloid/metabolism , Amyloid/toxicity , Amyloid beta-Peptides/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/pathology , Cells, Cultured , Hippocampus/cytology , Neurites/drug effects , Neurites/pathology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/therapeutic use , Peptide Fragments/chemical synthesis , Peptide Fragments/therapeutic use , PrPC Proteins/chemistry , Primary Cell Culture , Protein Aggregation, Pathological/pathology , Rats , Rats, Wistar
13.
BMC Neurosci ; 19(1): 7, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29471781

ABSTRACT

BACKGROUND: Neuroinflammation and the presence of amyloid beta protein (Aß) and neurofibrillary tangles are key pathologies in Alzheimer's disease (AD). As a potent anti-amyloid and anti-inflammatory natural polyphenol, curcumin (Cur) could be potential therapies for AD. Unfortunately, poor solubility, instability in physiological fluids, and low bioavailability limit its clinical utility. Recently, different lipid modifications in the formulae of Cur have been developed that would enhance its therapeutic potential. For example, we have reported greater permeability and neuroprotection with solid lipid curcumin particles (SLCP) than with natural Cur in an in vitro model of AD. In the present study, we compared the Aß aggregation inhibition, anti-amyloid, anti-inflammatory responses of Cur and or SLCP in both in vitro and in vivo models of AD. One-year-old 5xFAD-and age-matched wild-type mice were given intraperitoneal injections of Cur or SLCP (50 mg/kg body weight) for 2- or 5-days. Levels of Aß aggregation, including oligomers and fibril formation, were assessed by dot blot assay, while Aß plaque load and neuronal morphology in the pre-frontal cortex (PFC) and hippocampus were assayed by immunolabeling with Aß-specific antibody and cresyl violet staining, respectively. In addition, neuroinflammation was assessed the immunoreactivity (IR) of activated astrocytes (GFAP) and microglia (Iba-1) in different brain areas. Finally, comparisons of solubility and permeability of Cur and SLCP were made in cultured N2a cells and in primary hippocampal neurons derived from E16 pups of 5xFAD mice. RESULTS: We observed that relative to Cur, SLCP was more permeable, labeled Aß plaques more effectively, and produced a larger decrease in Aß plaque loads in PFC and dentate gyrus (DG) of hippocampus. Similarly, relative to Cur, SLCP produced a larger decrease of pyknotic, or tangle-like, neurons in PFC, CA1, and CA3 areas of hippocampus after 5 days of treatment. Both Cur and or SLCP significantly reduced GFAP-IR and Iba-1-IR in PFC, in the striatum as well as CA1, CA3, DG, subicular complex of hippocampus, and the entorhinal cortex in the 5xFAD mice after 5 days of treatment. CONCLUSIONS: The use of SLCP provides more anti-amyloid, anti-inflammatory, and neuroprotective outcomes than does Cur in the 5xFAD mouse model of AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Curcumin/pharmacology , Neuroprotective Agents/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Disease Models, Animal , Hippocampus/drug effects , Mice, Transgenic , Neurofibrillary Tangles/drug effects , Neurons/drug effects , Plaque, Amyloid/drug therapy
14.
Neural Plast ; 2018: 5257285, 2018.
Article in English | MEDLINE | ID: mdl-29755512

ABSTRACT

Alzheimer's disease (AD) is the most common type of dementia, mainly encompassing cognitive decline in subjects aged ≥65 years. Further, AD is characterized by selective synaptic and neuronal degeneration, vascular dysfunction, and two histopathological features: extracellular amyloid plaques composed of amyloid beta peptide (Aß) and neurofibrillary tangles formed by hyperphosphorylated tau protein. Dementia and AD are chronic neurodegenerative conditions with a complex physiopathology involving both genetic and environmental factors. Recent clinical studies have shown that proton pump inhibitors (PPIs) are associated with risk of dementia, including AD. However, a recent case-control study reported decreased risk of dementia. PPIs are a widely indicated class of drugs for gastric acid-related disorders, although most older adult users are not treated for the correct indication. Although neurological side effects secondary to PPIs are rare, several preclinical reports indicate that PPIs might increase Aß levels, interact with tau protein, and affect the neuronal microenvironment through several mechanisms. Considering the controversy between PPI use and dementia risk, as well as both cognitive and neuroprotective effects, the aim of this review is to examine the relationship between PPI use and brain effects from a neurobiological and clinical perspective.


Subject(s)
Dementia/chemically induced , Dementia/metabolism , Proton Pump Inhibitors/metabolism , Proton Pump Inhibitors/therapeutic use , Aged , Aged, 80 and over , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/drug effects , Brain/metabolism , Case-Control Studies , Dementia/drug therapy , Humans , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/metabolism , Plaque, Amyloid/chemically induced , Plaque, Amyloid/drug therapy , Plaque, Amyloid/metabolism , Proton Pump Inhibitors/adverse effects , tau Proteins/metabolism
15.
J Recept Signal Transduct Res ; 36(5): 445-58, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27305963

ABSTRACT

Glycogen synthase kinase-3ß (GSK-3ß) is a serine/threonine kinase which has attracted significant attention during recent years in drug design studies. The deregulation of GSK-3ß increased the loss of hippocampal neurons by triggering apoptosis-mediating production of neurofibrillary tangles and alleviates memory deficits in Alzheimer's disease (AD). Given its role in the formation of neurofibrillary tangles leading to AD, it has been a major therapeutic target for intervention in AD, hence was targeted in the present study. Twenty crystal structures were refined to generate pharmacophore models based on energy involvement in binding co-crystal ligands. Four common e-pharmacophore models were optimized from the 20 pharmacophore models. Shape-based screening of four e-pharmacophore models against nine established small molecule databases using Phase v3.9 had resulted in 1800 compounds having similar pharmacophore features. Rigid receptor docking (RRD) was performed for 1800 compounds and 20 co-crystal ligands with GSK-3ß to generate dock complexes. Interactions of the best scoring lead obtained through RRD were further studied with quantum polarized ligand docking (QPLD), induced fit docking (IFD) and molecular mechanics/generalized Born surface area. Comparing the obtained leads to 20 co-crystal ligands resulted in 18 leads among them, lead1 had the lowest docking score, lower binding free energy and better binding orientation toward GSK-3ß. The 50 ns MD simulations run confirmed the stable nature of GSK-3ß-lead1 docking complex. The results from RRD, QPLD, IFD and MD simulations confirmed that lead1 might be used as a potent antagonist for GSK-3ß.


Subject(s)
Alzheimer Disease/drug therapy , Glycogen Synthase Kinase 3 beta/chemistry , Protein Conformation , Small Molecule Libraries/chemistry , Alzheimer Disease/pathology , Apoptosis/drug effects , Crystallography, X-Ray , Drug Design , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Hippocampus/drug effects , Hippocampus/pathology , Humans , Ligands , Memory/drug effects , Molecular Docking Simulation , Neurofibrillary Tangles/chemistry , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/pathology , Neurons/drug effects , Neurons/pathology , Protein Binding , Small Molecule Libraries/therapeutic use , User-Computer Interface
16.
Neurochem Res ; 41(11): 3074-3082, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27518086

ABSTRACT

The pathological features of Alzheimer's disease (AD) include extracellular neuritic plaques containing ß-amyloid (Aß) peptide, a cleaved fragment of amyloid precursor protein (APP) via ß-site amyloid precursor protein-cleaving enzyme 1 (BACE1) and intracellular neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau. Cyclin-dependent kinase 5 (Cdk5) is increasingly thought to play a pivotal role in the pathogenesis of AD, both as a regulator of the production of Aß and through its well-established role as a tau kinase. Fuzhisan (FZS), a Chinese herbal complex prescription, has been used for the treatment AD for over 20 years, and is known to enhance the cognitive ability in AD patients as well as in AD model rats. To investigate mechanisms of AD and the potential therapy of FZS in AD, we treated senescence-accelerated mouse SAMP8 mice, a useful model of AD-related memory impairment, with FZS by intragastrical administration for 8 weeks and Donepizel was used as a positive control. The results showed that FZS (0.3, 0.6, and 1.2 g/kg/day) improved impaired cognitive ability of aged SAMP8 mice in a dose-dependent manner. FZS robustly decreased Aß level and phosphorylation of tau. This was accompanied by a significant decrease in the BACE1 level and phosphorylated APP (Thr668). Futhermore, The p25/Cdk5 pathway was markedly down-regulated by FZS treatment. These results indicated that the memory ameliorating effect of FZS may be, in part, by regulation the p25/Cdk5 pathway which may contribute to down-regulation of Aß and tau hyperphosphorylation.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/biosynthesis , Drugs, Chinese Herbal/pharmacology , Hippocampus/drug effects , Neurofibrillary Tangles/drug effects , tau Proteins/metabolism , Aging , Alzheimer Disease/metabolism , Animals , Cyclin-Dependent Kinase 5/metabolism , Disease Models, Animal , Down-Regulation , Hippocampus/metabolism , Male , Mice , Neurofibrillary Tangles/metabolism , Phosphorylation/drug effects
17.
Br J Nutr ; 115(3): 449-65, 2016 Feb 14.
Article in English | MEDLINE | ID: mdl-26652155

ABSTRACT

Curcumin derived from turmeric is well documented for its anti-carcinogenic, antioxidant and anti-inflammatory properties. Recent studies show that curcumin also possesses neuroprotective and cognitive-enhancing properties that may help delay or prevent neurodegenerative diseases, including Alzheimer's disease (AD). Currently, clinical diagnosis of AD is onerous, and it is primarily based on the exclusion of other causes of dementia. In addition, phase III clinical trials of potential treatments have mostly failed, leaving disease-modifying interventions elusive. AD can be characterised neuropathologically by the deposition of extracellular ß amyloid (Aß) plaques and intracellular accumulation of tau-containing neurofibrillary tangles. Disruptions in Aß metabolism/clearance contribute to AD pathogenesis. In vitro studies have shown that Aß metabolism is altered by curcumin, and animal studies report that curcumin may influence brain function and the development of dementia, because of its antioxidant and anti-inflammatory properties, as well as its ability to influence Aß metabolism. However, clinical studies of curcumin have revealed limited effects to date, most likely because of curcumin's relatively low solubility and bioavailability, and because of selection of cohorts with diagnosed AD, in whom there is already major neuropathology. However, the fresh approach of targeting early AD pathology (by treating healthy, pre-clinical and mild cognitive impairment-stage cohorts) combined with new curcumin formulations that increase bioavailability is renewing optimism concerning curcumin-based therapy. The aim of this paper is to review the current evidence supporting an association between curcumin and modulation of AD pathology, including in vitro and in vivo studies. We also review the use of curcumin in emerging retinal imaging technology, as a fluorochrome for AD diagnostics.


Subject(s)
Alzheimer Disease/diagnosis , Alzheimer Disease/prevention & control , Curcumin/pharmacology , Animals , Cognition/drug effects , Disease Models, Animal , Fluorescent Dyes/analysis , Humans , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/metabolism , Neuroprotective Agents/pharmacology , Nootropic Agents/pharmacology , Radioligand Assay/methods , Randomized Controlled Trials as Topic
18.
Bioorg Med Chem Lett ; 26(20): 5024-5028, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27624076

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia. In an AD patient's brain, senile plaques and neurofibrillary tangles, the abnormal aggregates of amyloid ß (Aß) peptide and tau protein, are observed as the two major hallmarks of this disease. To develop a new drug for treatment of AD, we have designed and synthesized a series of curcumin derivatives and evaluated their inhibitory activities against both tau and Aß aggregation. In this study, we describe the development of the more potent aggregation inhibitor 3-[(1E)-2-(1H-indol-6-yl)ethenyl]-5-[(1E)-2-[2-methoxy-4-(2-pyridylmethoxy) phenyl] ethenyl]-1H-pyrazole (compound 4, PE859). This compound has a better pharmacokinetic profile and pharmacological efficacy in vivo than curcumin, making it suitable as a drug for AD.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Curcumin/analogs & derivatives , Curcumin/pharmacology , tau Proteins/antagonists & inhibitors , Animals , Curcumin/chemical synthesis , Drug Design , Inhibitory Concentration 50 , Mice , Mice, Transgenic , Neurofibrillary Tangles/drug effects , Structure-Activity Relationship , tau Proteins/genetics
19.
J Pathol ; 235(5): 721-30, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25430817

ABSTRACT

Amyloid ß peptide (Aß) immunization of Alzheimer's disease (AD) patients has been reported to induce amyloid plaque removal, but with little impact on cognitive decline. We have explored the consequences of Aß immunotherapy on neurons in post mortem brain tissue. Eleven immunized (AN1792, Elan Pharmaceuticals) AD patients were compared to 28 non-immunized AD cases. Immunohistochemistry on sections of neocortex was performed for neuron-specific nuclear antigen (NeuN), neurofilament protein (NFP) and phosphorylated-(p)PKR (pro-apoptotic kinase detected in degenerating neurons). Quantification was performed for pPKR and status spongiosis (neuropil degeneration), NeuN-positive neurons/field, curvature of the neuronal processes and interneuronal distance. Data were corrected for age, gender, duration of dementia and APOE genotype and also assessed in relation to Aß42 and tau pathology and key features of AD. In non-immunized patients, the degree of neuritic curvature correlated with spongiosis and pPKR, and overall the neurodegenerative markers correlated better with tau pathology than Aß42 load. Following immunization, spongiosis increased, interneuronal distance increased, while the number of NeuN-positive neurons decreased, consistent with enhanced neuronal loss. However, neuritic curvature was reduced and pPKR was associated with Aß removal in immunized patients. In AD, associations of spongiosis status, curvature ratio and pPKR load with microglial markers Iba1, CD68 and CD32 suggest a role for microglia in neurodegeneration. After immunization, correlations were detected between the number of NeuN-positive neurons and pPKR with Iba1, CD68 and CD64, suggesting that microglia are involved in the neuronal loss. Our findings suggest that in established AD this form of active Aß immunization may predominantly accelerate loss of damaged degenerating neurons. This interpretation is consistent with in vivo imaging indicating an increased rate of cerebral atrophy in immunized AD patients.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Vaccines/therapeutic use , Amyloid beta-Peptides/therapeutic use , Neocortex/drug effects , Neurons/drug effects , Aged , Aged, 80 and over , Alzheimer Disease/diagnosis , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/analysis , Antigens, Nuclear/analysis , Autopsy , Biomarkers/analysis , Case-Control Studies , Female , Humans , Male , Middle Aged , Neocortex/chemistry , Neocortex/immunology , Neocortex/pathology , Nerve Degeneration , Nerve Tissue Proteins/analysis , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/immunology , Neurofibrillary Tangles/pathology , Neurofilament Proteins/analysis , Neurons/chemistry , Neurons/immunology , Neurons/pathology , Peptide Fragments/analysis , Phosphorylation , Plaque, Amyloid , Treatment Outcome , eIF-2 Kinase/analysis , tau Proteins/analysis
20.
Am J Pathol ; 184(2): 520-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24269557

ABSTRACT

Alzheimer disease (AD) is a progressive neurodegenerative disorder with associated memory loss, spatial disorientation, and other psychiatric problems. Cholinergic system dysfunction is an early and salient feature of AD, and enhancing cholinergic signaling with acetylcholinesterase inhibitors is currently the primary strategy for improving cognition. The beneficial effects of acetylcholinesterase inhibitors, however, are typically short-lived and accompanied by adverse effects. Recent evidence suggests that activating α7 nicotinic acetylcholine receptors (α7 nAChR) may facilitate the specific modulation of brain cholinergic signaling, leading to cognitive enhancement and possibly to amelioration of AD pathologic findings. In the present study, we determined the effect of long-term treatment with the selective α7 nAChR agonist A-582941 in aged 3xTg-AD mice with robust AD-like pathology, which is particularly significant not only because this is the only mouse model that co-develops amyloid plaques and neurofibrillary tangles but also because it enabled us to explore whether A-582941 is able to restore brain function after the severe damage associated with AD. Analysis of ß-amyloid deposits, tau phosphorylation, and inflammatory cells revealed that, overall, pathologic findings were unchanged. Rather, α7 nAChR activation induced expression of c-Fos and brain-derived neurotrophic factor and phosphorylation of cyclic adenosine monophosphate response element binding and neurotrophic tyrosine receptor kinase type 2. More important, A-582941 completely restored cognition in aged 3xTg-AD mice to the level of that in age-matched nontransgenic mice. These novel findings indicate that activating α7 nAChR is a promising treatment for cognitive impairment in AD.


Subject(s)
Aging/pathology , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Cognition/drug effects , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , alpha7 Nicotinic Acetylcholine Receptor/agonists , Alzheimer Disease/metabolism , Amyloid beta-Peptides , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain/physiopathology , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Memory/drug effects , Mice , Mice, Transgenic , Neurofibrillary Tangles/drug effects , Nootropic Agents/pharmacology , Phosphorylation/drug effects , Plaque, Amyloid/metabolism , Plaque, Amyloid/physiopathology , Pyridazines/pharmacology , Pyrroles/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL