Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nat Commun ; 13(1): 5451, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36114178

ABSTRACT

Alzheimer's disease (AD) is the consequence of neuronal death and brain atrophy associated with the aggregation of protein tau into fibrils. Thus disaggregation of tau fibrils could be a therapeutic approach to AD. The small molecule EGCG, abundant in green tea, has long been known to disaggregate tau and other amyloid fibrils, but EGCG has poor drug-like properties, failing to fully penetrate the brain. Here we have cryogenically trapped an intermediate of brain-extracted tau fibrils on the kinetic pathway to EGCG-induced disaggregation and have determined its cryoEM structure. The structure reveals that EGCG molecules stack in polar clefts between the paired helical protofilaments that pathologically define AD. Treating the EGCG binding position as a pharmacophore, we computationally screened thousands of drug-like compounds for compatibility for the pharmacophore, discovering several that experimentally disaggregate brain-derived tau fibrils in vitro. This work suggests the potential of structure-based, small-molecule drug discovery for amyloid diseases.


Subject(s)
Alzheimer Disease , Amyloidosis , tau Proteins , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid/chemistry , Amyloid/drug effects , Catechin/analogs & derivatives , Catechin/chemistry , Catechin/pharmacology , Cryoelectron Microscopy , Drug Evaluation, Preclinical/methods , Humans , Tea/chemistry , tau Proteins/chemistry , tau Proteins/drug effects , tau Proteins/metabolism
2.
Nutrients ; 14(2)2022 Jan 07.
Article in English | MEDLINE | ID: mdl-35057429

ABSTRACT

For thousands of years, mankind has been using plant extracts or plants themselves as medicinal herbs. Currently, there is a great deal of public interest in naturally occurring medicinal substances that are virtually non-toxic, readily available, and have an impact on well-being and health. It has been noted that dietary curcumin is one of the regulators that may positively influence changes in the brain after ischemia. Curcumin is a natural polyphenolic compound with pleiotropic biological properties. The observed death of pyramidal neurons in the CA1 region of the hippocampus and its atrophy are considered to be typical changes for post-ischemic brain neurodegeneration and for Alzheimer's disease. Additionally, it has been shown that one of the potential mechanisms of severe neuronal death is the accumulation of neurotoxic amyloid and dysfunctional tau protein after cerebral ischemia. Post-ischemic studies of human and animal brains have shown the presence of amyloid plaques and neurofibrillary tangles. The significant therapeutic feature of curcumin is that it can affect the aging-related cellular proteins, i.e., amyloid and tau protein, preventing their aggregation and insolubility after ischemia. Curcumin also decreases the neurotoxicity of amyloid and tau protein by affecting their structure. Studies in animal models of cerebral ischemia have shown that curcumin reduces infarct volume, brain edema, blood-brain barrier permeability, apoptosis, neuroinflammation, glutamate neurotoxicity, inhibits autophagy and oxidative stress, and improves neurological and behavioral deficits. The available data suggest that curcumin may be a new therapeutic substance in both regenerative medicine and the treatment of neurodegenerative disorders such as post-ischemic neurodegeneration.


Subject(s)
Alzheimer Disease/drug therapy , Brain Ischemia/complications , Curcumin/pharmacology , Neuroprotective Agents/pharmacology , Alzheimer Disease/etiology , Amyloid/drug effects , Amyloid/metabolism , Animals , Apoptosis/drug effects , Atrophy/etiology , Biological Availability , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain Edema/drug therapy , Brain Ischemia/drug therapy , Curcumin/chemistry , Curcumin/pharmacokinetics , Disease Models, Animal , Gastrointestinal Microbiome/physiology , Gerbillinae , Hippocampus/pathology , Humans , Mice , Neuroinflammatory Diseases/drug therapy , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Oxidative Stress/drug effects , Rats , tau Proteins/drug effects , tau Proteins/metabolism
3.
Am J Chin Med ; 49(6): 1399-1415, 2021.
Article in English | MEDLINE | ID: mdl-34137676

ABSTRACT

Diabetes is a group of metabolic disorders with an increased risk of developing cognitive impairment and dementia. The hippocampus in the forebrain contains an abundance of insulin receptors related to cognitive function and plays an important role in the pathophysiology of neurodegenerative disorders. Berberine from traditional Chinese medicine has been used to treat diabetes and diabetic cognitive impairment, although its related mechanisms are largely unknown. In this study, a STZ diabetes rat model feeding with a high-fat diet was used to test the effects of berberine compared with metformin. Oral glucose tolerance and hyperinsulinemic-euglycemic clamp were used for glucose metabolism and insulin resistance. The Morris water maze was used to observe the compound effects on cognitive impairment. Serum and hippocampal [Formula: see text]-amyloid peptide (A[Formula: see text], Tau and phosphorylated Tau protein deposition in the hippocampi were measured. The TUNEL assay was used to detect the neuronal apoptosis, supported by histomorphological changes and transmissional electron microscopy (TEM) image. Our data showed that the diabetic rats had a significantly cognitive impairment. In addition to improving glucose metabolism and reducing insulin resistance, berberine significantly improved the cognitive function in the rat. Berberine also effectively decreased the expression of hippocampal tau protein, phosphorylated Tau, and increased insulin receptor antibodies. Moreover, berberine downregulated the abnormal phosphorylation of A[Formula: see text] and Tau protein and improved hippocampal insulin signaling. The TUNEL assay confirmed that berberine reduced hippocampal neuronal apoptosis supported by TEM. Thus, berberine significantly improved the cognitive function in diabetic rats by changing the peripheral and central insulin resistance. The reduction of neuronal injury, A[Formula: see text] deposition, abnormal phosphorylation of Tau protein, and neuronal apoptosis in the hippocampus were observed as the related mechanisms of action.


Subject(s)
Berberine/pharmacology , Cognitive Dysfunction/drug therapy , Diabetes Mellitus, Experimental/drug therapy , Metformin/pharmacology , Animals , Apoptosis/drug effects , Diet, High-Fat , Disease Models, Animal , Hippocampus/drug effects , Hypoglycemic Agents/pharmacology , Insulin Resistance/physiology , Male , Rats , Rats, Sprague-Dawley , Streptozocin , tau Proteins/drug effects
4.
Neurosci Lett ; 759: 135981, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34023407

ABSTRACT

Alzheimer's disease perpetually demands enormous research on the development of effective treatment strategies. The present study aims to define the role of Oxyresveratrol (OXY) alone and in combination with Alkoxy glycerols (AKG) to reduce Tau protein level and improve the climbing behaviour of Drosophila fly models expressed with human-Tau protein. Oxyresveratrol, a polyphenolic stilbene, possesses a wide range of biological activities like antioxidant, anti-inflammatory, and neuroprotective effects. Nevertheless, chemical instability and low solubility of OXY in aqueous solutions reduce its bioavailability and hinder it from exerting neuroprotective activities. An inclusion complex of OXY with ß- cyclodextrin (CD) (OXY-CD complex) was employed in the study for increased dissolution rate and oral availability of OXY. Fish oils and their derivatives have a plethora of applications in in vivo biological activities. Herein, we also remark on the role of AKG in reducing Tau protein level in flies by enhancing OXY-CD activity. Dietary supplementation of OXY-CD together with AKG improved the learning and memory abilities during the climbing assay in Tau flies. The study highlights OXY-CD and AKG as neuroprotective agents and put forward a plausible approach towards the increased permeability of pharmacological agents across the blood-brain barrier (BBB) for the central nervous system elicited by AKG.


Subject(s)
Alzheimer Disease/pathology , Glycerol/pharmacology , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Stilbenes/pharmacology , tau Proteins/drug effects , Animals , Animals, Genetically Modified , Behavior, Animal/drug effects , Benzofurans/pharmacology , Drosophila melanogaster , Humans , Learning/drug effects , Memory/drug effects
5.
J Ethnopharmacol ; 269: 113725, 2021 Apr 06.
Article in English | MEDLINE | ID: mdl-33352241

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Ganoderma lucidum (G. lucidum, Lingzhi), also known as "immortality mushroom" has been broadly used to improve health and longevity for thousands of years in Asia. G. lucidum and its spores have been used to promote health, based on its broad pharmacological and therapeutic activity. This species is recorded in Chinese traditional formula as a nootropic and has been suggested to improve cognitive dysfunction in Alzheimer's disease. However, little is known about the nootropic effects and molecular mechanism of action of G. lucidum spores. AIM OF THE STUDY: The present study investigated the protective effects of sporoderm-deficient Ganoderma lucidum spores (RGLS) against learning and memory impairments and its mechanism of action. MATERIALS AND METHODS: In the Morris water maze, the effects of RGLS on learning and memory impairments were evaluated in a rat model of sporadic Alzheimer's disease that was induced by an intracerebroventricular injection of streptozotocin (STZ). Changes in amyloid ß (Aß) expression, Tau expression and phosphorylation, brain-derived neurotrophic factor (BDNF), and the BDNF receptor tropomyosin-related kinase B (TrkB) in the hippocampus were evaluated by Western blot. RESULTS: Treatment with RGLS (360 and 720 mg/kg) significantly enhanced memory in the rat model of STZ-induced sporadic Alzheimer's disease and reversed the STZ-induced increases in Aß expression and Tau protein expression and phosphorylation at Ser199, Ser202, and Ser396. The STZ-induced decreases in neurotrophic factors, including BDNF, TrkB and TrkB phosphorylation at Tyr816, were reversed by treatment with RGLS. CONCLUSION: These findings indicate that RGLS prevented learning and memory impairments in the present rat model of STZ-induced sporadic Alzheimer's disease, and these effects depended on a decrease in Aß expression and Tau hyperphosphorylation and the modulation of BDNF-TrkB signaling in the hippocampus.


Subject(s)
Alzheimer Disease/drug therapy , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Memory Disorders/prevention & control , Reishi/chemistry , Spores, Fungal/chemistry , Alzheimer Disease/chemically induced , Amyloid beta-Peptides/metabolism , Animals , Brain-Derived Neurotrophic Factor/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Drugs, Chinese Herbal/isolation & purification , Drugs, Chinese Herbal/therapeutic use , Hippocampus/drug effects , Male , Maze Learning/drug effects , Memory Disorders/chemically induced , Phosphorylation/drug effects , Plaque, Amyloid/chemically induced , Plaque, Amyloid/prevention & control , Rats, Sprague-Dawley , Receptor, trkB/drug effects , Receptor, trkB/metabolism , Signal Transduction/drug effects , Streptozocin/toxicity , tau Proteins/drug effects , tau Proteins/metabolism
6.
Phytother Res ; 34(11): 2867-2888, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32491273

ABSTRACT

Resveratrol (Res) is a non-flavonoid compound with pharmacological actions such as antioxidant, antiinflammatory, hepatoprotective, antidiabetes, and antitumor. This plant-derived chemical has a long history usage in treatment of diseases. The excellent therapeutic impacts of Res and its capability in penetration into blood-brain barrier have made it an appropriate candidate in the treatment of neurological disorders (NDs). Tau protein aggregations and amyloid-beta (Aß) deposits are responsible for the induction of NDs. A variety of studies have elucidated the role of these aggregations in NDs and the underlying molecular pathways in their development. In the present review, based on the recently published articles, we describe that how Res administration could inhibit amyloidogenic pathway and stimulate processes such as autophagy to degrade Aß aggregations. Besides, we demonstrate that Res supplementation is beneficial in dephosphorylation of tau proteins and suppressing their aggregations. Then, we discuss molecular pathways and relate them to the treatment of NDs.


Subject(s)
Amyloid beta-Peptides/drug effects , Resveratrol/therapeutic use , tau Proteins/drug effects , Humans , Resveratrol/pharmacology
7.
Phytother Res ; 34(4): 796-807, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31795012

ABSTRACT

Alzheimer's disease is a neurodegenerative disorder characterized by extracellular deposition of amyloid-ß (Aß) peptide and hyperphosphorylation of Tau protein, which ultimately leads to the formation of intracellular neurofibrillary tangles and cell death. Increasing evidence indicates that genistein, a soy isoflavone, has neuroprotective effects against Aß-induced toxicity. However, the molecular mechanisms involved in its neuroprotection are not well understood. In this study, we have established a neuronal damage model using retinoic-acid differentiated SH-SY5Y cells treated with different concentrations of Aß25-35 to investigate the effect of genistein against Aß-induced cell death and the possible involvement of protein kinase B (PKB, also termed Akt), glycogen synthase kinase 3ß (GSK-3ß), and Tau as an underlying mechanism to this neuroprotection. Differentiated SH-SY5Y cells were pre-treated for 24 hr with genistein (1 and 10 nM) and exposed to Aß25-35 (25 µM), and we found that genistein partially inhibited Aß induced cell death, primarily apoptosis. Furthermore, the protective effect of genistein was associated with the inhibition of Aß-induced Akt inactivation and Tau hyperphosphorylation. These findings reinforce the neuroprotective effects of genistein against Aß toxicity and provide evidence that its mechanism may involve regulation of Akt and Tau proteins.


Subject(s)
Amyloid beta-Peptides/toxicity , Genistein/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Apoptosis/drug effects , Cell Death/drug effects , Cell Line, Tumor , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Neurons/physiology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , tau Proteins/drug effects , tau Proteins/metabolism
8.
Int J Biol Macromol ; 120(Pt A): 1140-1148, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30179693

ABSTRACT

The application of nanomaterials in the healthy system may induce some neurodegenerative diseases initiated by tau folding and neuronal cell death. Herein, aluminium oxide nanoparticles (Al2O3 NPs) were synthesized and characterized by XRD, TEM, DLS and zeta potential investigations. Afterwards, the interaction of Al2O3 NPs with tau protein was investigated by fluorescence and CD spectroscopic methods. The molecular docking and molecular dynamic were also run to explore the binding site and conformational changes of tau after interaction with Al2O3 cluster. Moreover, the MTT, LDH, caspase-9/-3 and flow cytometry assays were done to explore the Al2O3 NPs-induced cytotoxicity against SH-SY5Y cells. It was revealed that Al2O3 NPs bind to tau protein and form a static complex and fold the structure of tau toward a more packed structure. Molecular docking and molecular dynamic investigations revealed that NPs bind to the hydrophilic residues of the tau segments and promote some marginal structural folding of tau segment. The cellular assays displayed that Al2O3 NPs can elicit cell mortality through membrane leakage, caspase-9/-3 activations, and induction of both apoptosis and necrosis. This data may indicate that NPs can induce some adverse effects on the biological systems.


Subject(s)
Aluminum Oxide/administration & dosage , Metal Nanoparticles/administration & dosage , Neuroblastoma/drug therapy , tau Proteins/chemistry , Aluminum Oxide/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Metal Nanoparticles/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Neuroblastoma/chemistry , Neuroblastoma/pathology , Protein Conformation/drug effects , tau Proteins/drug effects
9.
Sci Rep ; 8(1): 6431, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29691439

ABSTRACT

Accumulation of amyloid-ß plaques and tau contribute to the pathogenesis of Alzheimer's disease (AD), but it is unclear whether targeting tau pathology by antioxidants independently of amyloid-ß causes beneficial effects on memory and neuropsychiatric symptoms. Selenium, an essential antioxidant element reduced in the aging brain, prevents development of neuropathology in AD transgenic mice at early disease stages. The therapeutic potential of selenium for ameliorating or reversing neuropsychiatric and cognitive behavioral symptoms at late AD stages is largely unknown. Here, we evaluated the effects of chronic dietary sodium selenate supplementation for 4 months in female 3xTg-AD mice at 12-14 months of age. Chronic sodium selenate treatment efficiently reversed hippocampal-dependent learning and memory impairments, and behavior- and neuropsychiatric-like symptoms in old female 3xTg-AD mice. Selenium significantly decreased the number of aggregated tau-positive neurons and astrogliosis, without globally affecting amyloid plaques, in the hippocampus of 3xTg-AD mice. These results indicate that selenium treatment reverses AD-like memory and neuropsychiatric symptoms by a mechanism involving reduction of aggregated tau and/or reactive astrocytes but not amyloid pathology. These results suggest that sodium selenate could be part of a combined therapeutic approach for the treatment of memory and neuropsychiatric symptoms in advanced AD stages.


Subject(s)
Memory/drug effects , Selenic Acid/pharmacology , tau Proteins/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Dietary Supplements , Disease Models, Animal , Female , Hippocampus/metabolism , Humans , Maze Learning/drug effects , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Plaque, Amyloid/pathology , Presenilin-1/metabolism , Selenic Acid/metabolism , Selenium/metabolism , Selenium/pharmacology , tau Proteins/metabolism
10.
Neuroreport ; 28(16): 1043-1048, 2017 Nov 08.
Article in English | MEDLINE | ID: mdl-28902708

ABSTRACT

Tauopathies are neurodegenerative diseases that are characterized by the presence of hyperphosphorylated tau-containing neurofibrillary tangles (NFTs) in the brain and include Alzheimer's disease and frontotemporal dementia, which lack effective disease-modifying treatments. The presence of NFTs is known to correlate with cognition impairment, suggesting that targeting tau hyperphosphorylation may be therapeutically effective. MLC901 is a herbal formulation that is currently used in poststroke recovery and consists of nine herbal components. Previously, several components of MLC901 have been shown to have an effect on tau phosphorylation, but it remains unknown whether MLC901 itself has the same effect. The objective of this study was to assess the effects of MLC901 on ameliorating tau phosphorylation at epitopes associated with NFT formation. A stably transfected cell culture model expressing tau harboring the P301S mutation was generated and treated with various concentrations of MLC901 across different time points. Tau phosphorylation profiles and protein levels of enzymes associated with tau phosphorylation were assessed using western blotting. One-way analysis of variance with Bonferroni post-hoc analysis showed that MLC901 significantly reduced tau phosphorylation at epitopes recognized by the AT8, AT270, and PHF-13 antibodies. MLC901 also induced a significant increase in the s9 phosphorylation of glycogen synthase kinase 3ß and a concurrent decrease in the activation of cyclin-dependent kinase 5, as measured by a significant decrease in the levels of p35/cyclin-dependent kinase 5. Our results provide supporting evidence to further study the effects of MLC901 on tau pathology and cognition using mouse models of tauopathy.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Neurofibrillary Tangles/metabolism , Neuroprotective Agents/pharmacology , tau Proteins/drug effects , tau Proteins/metabolism , Cells, Cultured , Humans , Phosphorylation/drug effects
11.
ACS Chem Neurosci ; 7(7): 912-23, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27213824

ABSTRACT

Alzheimer's disease (AD) is the most common brain disorder worldwide. Aberrant tau hyperphosphorylation and accumulation play critical roles in the formation of neurofibrillary tangles highly associated with neuronal dysfunction and cognitive impairment in AD pathogenesis. Glycogen synthase kinase-3ß (GSK3ß) is a key kinase responsible for tau hyperphosphorylation. Selective inhibition of GSK3ß is a promising strategy in AD therapy. Corn silks (CS, Zea mays L.) have been traditionally used as a medicinal herb and recently noted for their potentially cognitive benefits. However, the neuroprotective components of CS and their molecular mechanism have received little attention to date. As part of our effort screening phytochemicals against a broad panel of kinases targeting AD tauopathy, we found inhibition of GSK3ß by CS extracts. Subsequent bioassay-guided fractionation led to the isolation and identification of two 6-C-glycosylflavones, isoorientin (1) and 3'-methoxymaysin (2), with selective inhibition against GSK3ß in vitro. Enzyme kinetics and molecular docking studies demonstrated that 1 specifically inhibited GSK3ß via an ATP noncompetitive mechanism, acting as a substrate competitive inhibitor of GSK3ß. Further in vitro cellular studies demonstrated that 1 effectively attenuated tau phosphorylation mediated by GSK3ß and was neuroprotective against ß-amyloid-induced tau hyperphosphorylation and neurotoxicity in SH-SY5Y cells. The C-glycosylflavones represent new lead candidates with a novel mechanism of action for the development of AD phytopharmaceuticals.


Subject(s)
Amyloid beta-Peptides/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/chemistry , Flavonoids/pharmacology , Glycogen Synthase Kinase 3/metabolism , Peptide Fragments/pharmacology , tau Proteins/metabolism , Adenosine Triphosphate/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Humans , Isoflavones/chemistry , Isoflavones/pharmacology , Luteolin/chemistry , Luteolin/pharmacology , Models, Chemical , Neuroblastoma/pathology , Phosphorylation/drug effects , Signal Transduction/drug effects , Staurosporine/pharmacology , tau Proteins/drug effects
12.
Brain ; 139(Pt 5): 1568-86, 2016 05.
Article in English | MEDLINE | ID: mdl-27020329

ABSTRACT

Amyloid-ß, tau, and α-synuclein, or more specifically their soluble oligomers, are the aetiologic molecules in Alzheimer's disease, tauopathies, and α-synucleinopathies, respectively. These proteins have been shown to interact to accelerate each other's pathology. Clinical studies of amyloid-ß-targeting therapies in Alzheimer's disease have revealed that the treatments after disease onset have little benefit on patient cognition. These findings prompted us to explore a preventive medicine which is orally available, has few adverse effects, and is effective at reducing neurotoxic oligomers with a broad spectrum. We initially tested five candidate compounds: rifampicin, curcumin, epigallocatechin-3-gallate, myricetin, and scyllo-inositol, in cells expressing amyloid precursor protein (APP) with the Osaka (E693Δ) mutation, which promotes amyloid-ß oligomerization. Among these compounds, rifampicin, a well-known antibiotic, showed the strongest activities against the accumulation and toxicity (i.e. cytochrome c release from mitochondria) of intracellular amyloid-ß oligomers. Under cell-free conditions, rifampicin inhibited oligomer formation of amyloid-ß, tau, and α-synuclein, indicating its broad spectrum. The inhibitory effects of rifampicin against amyloid-ß and tau oligomers were evaluated in APPOSK mice (amyloid-ß oligomer model), Tg2576 mice (Alzheimer's disease model), and tau609 mice (tauopathy model). When orally administered to 17-month-old APPOSK mice at 0.5 and 1 mg/day for 1 month, rifampicin reduced the accumulation of amyloid-ß oligomers as well as tau hyperphosphorylation, synapse loss, and microglial activation in a dose-dependent manner. In the Morris water maze, rifampicin at 1 mg/day improved memory of the mice to a level similar to that in non-transgenic littermates. Rifampicin also inhibited cytochrome c release from the mitochondria and caspase 3 activation in the hippocampus. In 13-month-old Tg2576 mice, oral rifampicin at 0.5 mg/day for 1 month decreased amyloid-ß oligomer accumulation, tau hyperphosphorylation, synapse loss, and microglial activation, but not amyloid deposition. Rifampicin treatment to 14-15-month-old tau609 mice at 0.5 and 1 mg/day for 1 month also reduced tau oligomer accumulation, tau hyperphosphorylation, synapse loss, and microglial activation in a dose-dependent fashion, and improved the memory almost completely at 1 mg/day. In addition, rifampicin decreased the level of p62/sequestosome-1 in the brain without affecting the increased levels of LC3 (microtubule-associated protein light chain 3) conversion, suggesting the restoration of autophagy-lysosomal function. Considering its prescribed dose and safety in humans, these results indicate that rifampicin could be a promising, ready-to-use medicine for the prevention of Alzheimer's disease and other neurodegenerative diseases.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/drug effects , Rifampin/pharmacology , Rifampin/therapeutic use , Tauopathies/prevention & control , tau Proteins/drug effects , Alzheimer Disease/complications , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Caspase 3/metabolism , Cells, Cultured , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Female , Hippocampus/metabolism , Maze Learning/drug effects , Memory Disorders/complications , Memory Disorders/drug therapy , Mice , Mice, Transgenic , Microglia/drug effects , Microtubule-Associated Proteins/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Phosphorylation/drug effects , Sequestosome-1 Protein/metabolism , Synapses/drug effects , Synucleins/drug effects , Synucleins/metabolism , Tauopathies/complications , Tauopathies/metabolism , tau Proteins/metabolism
13.
J Biomol Screen ; 21(8): 804-15, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26984927

ABSTRACT

Tau aggregation is the pathological hallmark that best correlates with the progression of Alzheimer's disease (AD). The presence of neurofibrillary tangles (NFTs), formed of hyperphosphorylated tau, leads to neuronal dysfunction and loss, and is directly associated with the cognitive decline observed in AD patients. The limited success in targeting ß-amyloid pathologies has reinforced the hypothesis of blocking tau phosphorylation, aggregation, and/or spreading as alternative therapeutic entry points to treat AD. Identification of novel therapies requires disease-relevant and scalable assays capable of reproducing key features of the pathology in an in vitro setting. Here we use induced pluripotent stem cells (iPSCs) as a virtually unlimited source of human cortical neurons to develop a robust and scalable tau aggregation model compatible with high-throughput screening (HTS). We downscaled cell culture conditions to 384-well plate format and used Matrigel to introduce an extra physical protection against cell detachment that reduces shearing stress and better recapitulates pathological conditions. We complemented the assay with AlphaLISA technology for the detection of tau aggregates in a high-throughput-compatible format. The assay is reproducible across users and works with different commercially available iPSC lines, representing a highly translational tool for the identification of novel treatments against tauopathies, including AD.


Subject(s)
Cell Culture Techniques/methods , High-Throughput Screening Assays/methods , Tauopathies/drug therapy , tau Proteins/chemistry , Brain/metabolism , Brain/pathology , Drug Evaluation, Preclinical/methods , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Phosphorylation , Protein Aggregates/genetics , Tauopathies/genetics , tau Proteins/drug effects , tau Proteins/genetics
14.
Nat Commun ; 6: 10216, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26671725

ABSTRACT

Neurofibrillary tangles, composed of hyperphosphorylated tau fibrils, are a pathological hallmark of Alzheimer's disease; the neurofibrillary tangle load correlates strongly with clinical progression of the disease. A growing body of evidence indicates that tau oligomer formation precedes the appearance of neurofibrillary tangles and contributes to neuronal loss. Here we show that tau oligomer formation can be inhibited by compounds whose chemical backbone includes 1,2-dihydroxybenzene. Specifically, we demonstrate that 1,2-dihydroxybenzene-containing compounds bind to and cap cysteine residues of tau and prevent its aggregation by hindering interactions between tau molecules. Further, we show that orally administered DL-isoproterenol, an adrenergic receptor agonist whose skeleton includes 1,2-dihydroxybenzene and which penetrates the brain, reduces the levels of detergent-insoluble tau, neuronal loss and reverses neurofibrillary tangle-associated brain dysfunction. Thus, compounds that target the cysteine residues of tau may prove useful in halting the progression of Alzheimer's disease and other tauopathies.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Alzheimer Disease/metabolism , Catechols/pharmacology , Cysteine/drug effects , Isoproterenol/pharmacology , Neurofibrillary Tangles/drug effects , Neurons/drug effects , tau Proteins/drug effects , Adrenergic beta-Agonists/chemistry , Animals , Behavior, Animal/drug effects , Blotting, Western , Brain/drug effects , Brain/metabolism , Catechols/chemistry , Catechols/metabolism , Cell Line, Tumor , Cysteine/metabolism , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical , Isoproterenol/chemistry , Mice , Mice, Transgenic , Neurofibrillary Tangles/metabolism , Neurons/pathology , Polymerization , tau Proteins/genetics , tau Proteins/metabolism
15.
Alzheimers Dement ; 9(4): 452-458.e1, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23809366

ABSTRACT

For decades, researchers have focused primarily on a pathway initiated by amyloid beta aggregation, amyloid deposition, and accumulation in the brain as the key mechanism underlying the disease and the most important treatment target. However, evidence increasingly suggests that amyloid is deposited early during the course of disease, even prior to the onset of clinical symptoms. Thus, targeting amyloid in patients with mild to moderate Alzheimer's disease (AD), as past failed clinical trials have done, may be insufficient to halt further disease progression. Scientists are investigating other molecular and cellular pathways and processes that contribute to AD pathogenesis. Thus, the Alzheimer's Association's Research Roundtable convened a meeting in April 2012 to move beyond amyloid and explore AD as a complex multifactorial disease, with the goal of using a more inclusive perspective to identify novel treatment strategies.


Subject(s)
Alzheimer Disease/drug therapy , Molecular Targeted Therapy , Nootropic Agents/therapeutic use , Aging , Alzheimer Disease/diagnosis , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Autophagy/drug effects , Biomarkers , Brain/metabolism , Cell Cycle/drug effects , Cooperative Behavior , Diabetes Mellitus, Type 2/complications , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Inflammation , Insulin Resistance , Lysosomes/drug effects , Lysosomes/physiology , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/pathology , Models, Neurological , Neuroimaging , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nootropic Agents/pharmacology , Public-Private Sector Partnerships , Resource Allocation , tau Proteins/drug effects , tau Proteins/physiology
16.
J Alzheimers Dis ; 36(1): 21-40, 2013.
Article in English | MEDLINE | ID: mdl-23531502

ABSTRACT

Abnormal modifications in tau such as hyperphosphorylation, oxidation, and glycation interfere with its interaction with microtubules leading to its dissociation and self-aggregation into neurofibrillary tangles, a hallmark of Alzheimer's disease (AD). Previously we reported that an aqueous extract of cinnamon has the ability to inhibit tau aggregation in vitro and can even induce dissociation of tangles isolated from AD brain. In the present study, we carried out investigations with cinnamaldehyde (CA) and epicatechin (EC), two components of active cinnamon extract. We found that CA and the oxidized form of EC (ECox) inhibited tau aggregation in vitro and the activity was due to their interaction with the two cysteine residues in tau. Mass spectrometry of a synthetic peptide, SKCGS, representing the actual tau sequence, identified the thiol as reacting with CA and ECox. Use of a cysteine double mutant of tau showed the necessity of cysteine for aggregation inhibition by CA. The interaction of CA with tau cysteines was reversible and the presence of CA did not impair the biological function of tau in tubulin assembly in vitro. Further, these compounds protected tau from oxidation caused by the reactive oxygen species, H2O2, and prevented subsequent formation of high molecular weight species that are considered to stimulate tangle formation. Finally, we observed that EC can sequester highly reactive and toxic byproducts of oxidation such as acrolein. Our results suggest that small molecules that form a reversible interaction with cysteines have the potential to protect tau from abnormal modifications.


Subject(s)
Acrolein/analogs & derivatives , Alzheimer Disease/drug therapy , Catechin/therapeutic use , tau Proteins/drug effects , Acrolein/therapeutic use , Alzheimer Disease/etiology , Animals , Cattle , Cysteine/metabolism , In Vitro Techniques , Microscopy, Electron, Transmission , Microtubules/metabolism , Microtubules/ultrastructure , Oxidation-Reduction , Spectrometry, Mass, Electrospray Ionization , Tubulin/biosynthesis , Tubulin/ultrastructure , tau Proteins/metabolism
17.
J Nat Prod ; 75(9): 1584-8, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22988908

ABSTRACT

Among the phenolic compounds extracted from extra virgin olive oil, oleocanthal (1) has attracted considerable attention in the modulation of many human diseases, such as inflammation and Alzheimer's disease (AD). Indeed, 1 is capable of altering the fibrillization of tau protein, which is one of the key factors at the basis of neurodegenerative diseases, and of covalently reacting with lysine ε-amino groups of the tau fragment K18 in an unspecific fashion. In the present study, an investigation of the recognition process and the reaction profile between 1 and the wild-type tau protein has been conducted by a circular dichroism, surface plasmon resonance, fluorescence, and mass spectrometry combined approach. As a result, 1 has been found to interact with tau-441, inducing stable conformational modifications of the protein secondary structure and also interfering with tau aggregation. These findings provide experimental support for the potential reduced risk of AD and related neurodegenerative diseases associated with olive oil consumption and may offer a new chemical scaffold for the development of AD-modulating agents.


Subject(s)
Aldehydes/chemistry , Aldehydes/pharmacology , Phenols/chemistry , Phenols/pharmacology , tau Proteins/drug effects , Alzheimer Disease/metabolism , Cyclopentane Monoterpenes , Humans , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Olive Oil , Plant Oils/chemistry , tau Proteins/chemistry , tau Proteins/metabolism
18.
Planta Med ; 78(12): 1337-41, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22700047

ABSTRACT

Alzheimer's disease is the most common form of dementia leading to the irreversible loss of neurons, and Tau hyperphosphorylation has an important role in the pathology of Alzheimer's disease. Ginkgolide A is one of the active components of Ginkgo biloba extracts which has been proven to have neuroprotective effects, but the effect of ginkgolide A on Tau hyperphosphorylation has not yet been reported. In this study, the effects of ginkgolide A on cell viability, Tau hyperphosphorylation, and the PI3K-Akt signaling pathway in N2a cell lines were explored, and methods such as the MTT assay, ELISA, and Western blots techniques were used. The results showed that ginkgolide A could increase cell viability and suppress the phosphorylation level of Tau in cell lysates, meanwhile, GSK3ß was inhibited with phosphorylation at Ser9. Moreover, treatment of the cells with ginkgolide A promoted phosphorylation of PI3K and Akt, suggesting that the activation of the PI3K-Akt signaling pathway may be the mechanism for ginkgolide A to prevent the intracellular accumulation of p-Tau induced by okadaic acid and to protect the cells from Tau hyperphosphorylation-related toxicity.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Ginkgo biloba/chemistry , Ginkgolides/pharmacology , Lactones/pharmacology , Phosphatidylinositol 3-Kinases/drug effects , tau Proteins/drug effects , Alzheimer Disease/metabolism , Animals , Cell Line/drug effects , Cell Survival/drug effects , Mice , Neuroblastoma , Okadaic Acid , Phosphorylation/drug effects , Plant Extracts/pharmacology , Signal Transduction/drug effects
19.
J Am Osteopath Assoc ; 110(9 Suppl 8): S27-36, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20926740

ABSTRACT

Development of effective treatments for patients with Alzheimer disease has been challenging. Currently approved treatments include acetylcholinesterase inhibitors and the N-methyl-D-aspartate receptor antagonist memantine hydrochloride. To investigate treatments in development for patients with Alzheimer disease, the author conducted a review of the literature. New approaches for treatment or prevention focus on several general areas, including cholinergic receptor agonists, drugs to decrease ß-amyloid and tau levels, antiinflammatory agents, drugs to increase nitric oxide and cyclic guanosine monophosphate levels, and substances to reduce cell death or promote cellular regeneration. The author focuses on medications currently in clinical trials. Cholinergic agents include orthostatic and allosteric muscarinic M1 agonists and nicotinic receptor agonists. Investigational agents that target ß-amyloid include vaccines, antibodies, and inhibitors of ß-amyloid production. Anti-inflammatory agents, including nonsteroidal anti-inflammatory drugs, the natural product curcumin, and the tumor necrosis factor α inhibitor etanercept, have also been studied. Some drugs currently approved for other uses may also show promise for treatment of patients with Alzheimer disease. Results of clinical trials with many of these investigational drugs have been disappointing, perhaps because of their use with patients in advanced stages of Alzheimer disease. Effective treatment may need to begin earlier-before neurodegeneration becomes severe enough for symptoms to appear.


Subject(s)
Alzheimer Disease/drug therapy , Drugs, Investigational , Alzheimer Vaccines , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/drug effects , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antibodies, Monoclonal/therapeutic use , Cholinergic Agents/therapeutic use , Etanercept , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Immunoglobulin G/therapeutic use , Indoles/therapeutic use , Nicotinic Antagonists/therapeutic use , Phosphodiesterase 5 Inhibitors/therapeutic use , Receptors, Tumor Necrosis Factor/therapeutic use , tau Proteins/drug effects
20.
Curr Alzheimer Res ; 7(3): 235-40, 2010 May.
Article in English | MEDLINE | ID: mdl-20088807

ABSTRACT

Tau aggregation is an appealing target for therapeutic intervention. However, conformational change or aggregation needs to be targeted without inhibiting the normal biology of tau and its role in microtubule stabilization. The number of compound classes being tested at this time are very limited and include Congo red derivatives [2], anthraquinones (Pickhardt et al. 2005 [3], disputed in Crowe et al. 2007 [4]), 2,3-di(furan-2-yl)-quinoxalines , phenylthiazolyl-hydrazide (PTH) [5], polyphenols and porphyrins [6] and cyanine dyes [1, 7, 8]. Herein we have utilized a member of the cyanine dye family (C11) in an organotypic slice culture model of tangle formation. Our results demonstrate that C11 is capable of affecting tau polymerization in a biphasic, dose dependent manner. At submicromolar concentrations (0.001 microM) C11 reduced levels of aggregated tau. However, higher doses resulted in an increase in tau polymerization. These effects can also be seen at the level of individual filaments with changes in filament length and number mirroring the pattern seen via immunoblotting. In addition, this effect is achieved without altering levels of phosphorylation at disease and microtubule binding relevant epitopes.


Subject(s)
Carbocyanines/pharmacology , Neurofibrillary Tangles/drug effects , Neurofibrillary Tangles/metabolism , Tauopathies/drug therapy , Tauopathies/metabolism , tau Proteins/drug effects , tau Proteins/metabolism , Animals , Binding Sites/drug effects , Binding Sites/physiology , Carbocyanines/therapeutic use , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Mice , Microtubules/drug effects , Microtubules/metabolism , Organ Culture Techniques , Polymers/chemistry , Structure-Activity Relationship , Tauopathies/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL