Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Cell Biochem ; 117(10): 2241-8, 2016 10.
Article in English | MEDLINE | ID: mdl-26910498

ABSTRACT

The objective of this study was to develop an in silico screening model for characterization of potential novel ligands from commercial drug libraries able to functionally activate certain olfactory receptors (ORs), which are members of the class A rhodopsin-like family of G protein couple receptors (GPCRs), in the brain of murine models of concussion. We previously found that concussions may significantly influence expression of certain ORs, for example, OR4M1 in subjects with a history of concussion/traumatic brain injury (TBI). In this study, we built a 3-D OR4M1 model and used it in in silico screening of potential novel ligands from commercial drug libraries. We report that in vitro activation of OR4M1 with the commercially available ZINC library compound 10915775 led to a significant attenuation of abnormal tau phosphorylation in embryonic cortico-hippocampal neuronal cultures derived from NSE-OR4M1 transgenic mice, possibly through modulation of the JNK signaling pathway. The attenuation of abnormal tau phosphorylation was rather selective since ZINC10915775 significantly decreased tau phosphorylation on tau Ser202/T205 (AT8 epitope) and tau Thr212/Ser214 (AT100 epitope), but not on tau Ser396/404 (PHF-1 epitope). Moreover, no response of ZINC10915775 was found in control hippocampal neuronal cultures derived from wild type littermates. Our in silico model provides novel means to pharmacologically modulate select ubiquitously expressed ORs in the brain through high affinity ligand activation to prevent and eventually to treat concussion induced down regulation of ORs and subsequent cascade of tau pathology. J. Cell. Biochem. 117: 2241-2248, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Brain Concussion/complications , Drug Discovery/methods , Pharmaceutical Preparations/metabolism , Receptors, Odorant/chemistry , Receptors, Odorant/metabolism , Tauopathies/drug therapy , tau Proteins/metabolism , Animals , Cells, Cultured , Computer Simulation , Epitopes , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Molecular , Molecular Docking Simulation , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Phosphorylation , Rats , Rats, Long-Evans , Tauopathies/etiology , Tauopathies/pathology
2.
J Alzheimers Dis ; 16(2): 433-9, 2009.
Article in English | MEDLINE | ID: mdl-19221432

ABSTRACT

Abnormal misfoldings of the microtubule-associated protein tau, leading to the aggregation of tau into paired helical filaments that are ultimately deposited as neurofibrillary tangles, is a key neuropathologic feature of a number of neurodegenerative disorders collectively referred to as tauopathies. We recently observed that a particular grape seed polyphenolic extract (GSPE), namely, Meganatural-Az may attenuate the generation and stability of misfolded proteins. We hypothesized that Meganatural-Az GSPE might also attenuate tau protein misfolding that leads to the generation of tau filamentary aggregates that are critical for the initiation and progression of neurodegeneration and/or cognitive dysfunctions in tauopathies. In this study, we used in vitro aggregations of synthetic Ac(306)VQIVYK(311) tau peptide as a model system to explore whether Meganatural-Az GSPE might modulate aggregations of tau protein. We demonstrate that this GSPE is capable of inhibiting tau peptide aggregations, as well as dissociating preformed tau peptide aggregates. Results from this study suggest that this GSPE might provide beneficial disease-modifying bioactivities in tau-associated neurodegenerative disorders by modulating tau-mediated neuropathologic mechanisms. Our observation, in conjunction with the demonstrated bioavailability, as well as safety and tolerability, of this GSPE, supports the development of Meganatural-Az GSPE for the prevention and/or treatment of tau-associated neurodegenerative disorders.


Subject(s)
Antioxidants/metabolism , Flavonoids/metabolism , Phenols/metabolism , Plant Extracts/chemistry , Tauopathies/metabolism , Antioxidants/pharmacology , Dose-Response Relationship, Drug , Flavonoids/pharmacology , Models, Biological , Peptides/metabolism , Phenols/pharmacology , Polyphenols , Protein Binding/drug effects , Protein Folding/drug effects
3.
Nat Commun ; 9(1): 477, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29396460

ABSTRACT

Major depressive disorder is associated with abnormalities in the brain and the immune system. Chronic stress in animals showed that epigenetic and inflammatory mechanisms play important roles in mediating resilience and susceptibility to depression. Here, through a high-throughput screening, we identify two phytochemicals, dihydrocaffeic acid (DHCA) and malvidin-3'-O-glucoside (Mal-gluc) that are effective in promoting resilience against stress by modulating brain synaptic plasticity and peripheral inflammation. DHCA/Mal-gluc also significantly reduces depression-like phenotypes in a mouse model of increased systemic inflammation induced by transplantation of hematopoietic progenitor cells from stress-susceptible mice. DHCA reduces pro-inflammatory interleukin 6 (IL-6) generations by inhibiting DNA methylation at the CpG-rich IL-6 sequences introns 1 and 3, while Mal-gluc modulates synaptic plasticity by increasing histone acetylation of the regulatory sequences of the Rac1 gene. Peripheral inflammation and synaptic maladaptation are in line with newly hypothesized clinical intervention targets for depression that are not addressed by currently available antidepressants.


Subject(s)
Anthocyanins/pharmacology , Caffeic Acids/pharmacology , Epigenesis, Genetic , Glucosides/pharmacology , Inflammation/genetics , Neuronal Plasticity/genetics , Stress, Psychological/genetics , Animals , Anthocyanins/administration & dosage , Caffeic Acids/administration & dosage , CpG Islands/drug effects , Depression/drug therapy , Drug Evaluation, Preclinical/methods , Glucosides/administration & dosage , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Leukocyte Common Antigens/genetics , Male , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Neuropeptides/genetics , Neuropeptides/metabolism , Polyphenols/pharmacology , Social Behavior , Stress, Psychological/drug therapy , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
4.
J Alzheimers Dis ; 54(2): 477-96, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27567821

ABSTRACT

The release of paired helical filaments (PHFs) from neurons into the extracellular space may contribute to the propagation of tau pathology across brain regions in Alzheimer's disease (AD) and other tauopathies. The majority of available mechanistic studies exploring the pathologic role of extracellular PHFs are conducted in proliferating cell lines. Here, we compare how extracellular PHFs induce tauopathy in mitotic cells and in post-mitotic brain neurons. In a mitotic cell line (HEK 293T), extracellular exposure to AD PHFs leads to an intracellular "aggresomal" type deposition of tau, coincidental with redistribution of dynein, a retrograde motor protein. We also observed that PHFs impaired proteasome degradation, but not autophagy. Exposure of cells to proteasome inhibitors was sufficient to induce intracellular tau aggregate formation as well as reorganization of dynein and the intermediate filament protein, vimentin. Thus, in mitotic cells, extracellular PHFs promote cellular tau aggregation, in part, by interfering with cellular proteasome degradation processes. In contrast with our observations with proliferating cells, exposure of post-mitotic primary neuronal cultures to AD PHFs did not promote "aggresomal" tau deposition, but instead resulted in a widespread accumulation of phosphorylated tau-immunoreactive swellings in neuritic processes, characterized by disturbed cytoskeletal organization of dynein and vimentin. Collectively, our observations suggest that extracellular PHFs may contribute to the propagation of tau pathology by independent mechanisms in post-mitotic and mitotic brain cells. These outcomes indicate that in addition to post-mitotic brain neurons, mitotic brain cells should also be considered as targets for therapeutic interventions to attenuate propagation of tauopathy.


Subject(s)
Brain/metabolism , Extracellular Fluid/metabolism , Mitosis/physiology , Neurofibrillary Tangles/metabolism , tau Proteins/metabolism , Aged , Aged, 80 and over , Animals , Brain/pathology , Cell Line, Tumor , Female , HEK293 Cells , Humans , Male , Mice , Neurofibrillary Tangles/pathology
5.
Neuromolecular Med ; 18(2): 177-89, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26984114

ABSTRACT

Caregiving for a dementia patient is associated with increased risk of psychological and physical health problems. We investigated whether a mindfulness-based stress reduction (MBSR) training course for caregivers that closely models the MBSR curriculum originally established by the Center of Mindfulness at the University of Massachusetts may improve the psychological resilience of non-professional caregivers of Alzheimer's disease patients. Twenty adult non-professional caregivers of dementia patients participated in an 8-week MBSR training course. Caregiver stress, depression, burden, grief, and gene expression profiles of blood mononuclear cells were assessed at baseline and following MBSR. MBSR training significantly improved the psychological resilience of some of the caregivers. We identified predictive biomarkers whose expression is associated with the likelihood of caregivers to benefit from MBSR, and biomarkers whose expression is associated with MBSR psychological benefits. Our biomarker studies provide insight into the mechanisms of health benefits of MBSR and a basis for developing a personalized medicine approach for applying MBSR for promoting psychological and cognitive resilience in caregivers of dementia patients.


Subject(s)
Biomarkers/blood , Caregivers/education , Caregivers/psychology , Mindfulness , Stress, Psychological/blood , Stress, Psychological/genetics , Gene Expression Regulation , Humans , Mindfulness/education , Mindfulness/standards , Stress, Psychological/diagnosis , Transcriptome
6.
Neurochem Int ; 89: 191-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26235983

ABSTRACT

Sleep deprivation produces deficits in hippocampal synaptic plasticity and hippocampal-dependent memory storage. Recent evidence suggests that sleep deprivation disrupts memory consolidation through multiple mechanisms, including the down-regulation of the cAMP-response element-binding protein (CREB) and of mammalian target of rapamycin (mTOR) signaling. In this study, we tested the effects of a Bioactive Dietary Polyphenol Preparation (BDPP), comprised of grape seed polyphenol extract, Concord grape juice, and resveratrol, on the attenuation of sleep deprivation-induced cognitive impairment. We found that BDPP significantly improves sleep deprivation-induced contextual memory deficits, possibly through the activation of CREB and mTOR signaling pathways. We also identified brain-available polyphenol metabolites from BDPP, among which quercetin-3-O-glucuronide activates CREB signaling and malvidin-3-O-glucoside activates mTOR signaling. In combination, quercetin and malvidin-glucoside significantly attenuated sleep deprivation-induced cognitive impairment in -a mouse model of acute sleep deprivation. Our data suggests the feasibility of using select brain-targeting polyphenol compounds derived from BDPP as potential therapeutic agents in promoting resilience against sleep deprivation-induced cognitive dysfunction.


Subject(s)
Brain/drug effects , Cognition Disorders/drug therapy , Grape Seed Extract/administration & dosage , Polyphenols/administration & dosage , Sleep Deprivation/drug therapy , Stilbenes/administration & dosage , Animals , Brain/metabolism , Cognition Disorders/etiology , Cognition Disorders/metabolism , Dose-Response Relationship, Drug , Drug Delivery Systems , Grape Seed Extract/metabolism , Mice , Mice, Inbred C57BL , Polyphenols/metabolism , Rats , Rats, Sprague-Dawley , Resveratrol , Sleep Deprivation/complications , Sleep Deprivation/metabolism , Stilbenes/metabolism
7.
Arch Neurol ; 60(3): 369-76, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12633148

ABSTRACT

BACKGROUND: Caspase gene expression has previously been reported in terminal Alzheimer disease (AD) brain, but, currently, little is known about the temporal pattern of caspase gene expression relative to the onset and clinical progression of AD. OBJECTIVE: To derive a profile of caspase gene expression and proapoptotic indexes as a function of the clinical and neuropathologic progression of AD dementia. SETTING AND PATIENTS: Postmortem survey of nursing home patients characterized clinically by Clinical Dementia Rating (CDR) and neuropathologically by Consortium to Establish a Registry for Alzheimer's Disease criteria. DESIGN AND OUTCOME MEASURES: To assess messenger RNA expression of caspase-1, -2L, -2S, -3, -5, -6, -7, -8, and -9; apoptotic cell death by TUNEL assay; and poly (ADP-ribose) polymerase cleavage in postmortem brain tissue samples from cognitively normal (CDR 0), high risk of developing AD dementia (CDR 0.5), and severe dementia (CDR 5) cases. RESULTS: Compared with CDR 0 cases, elevated messenger RNA expression of caspase-1 and caspase-7 in the entorhinal cortex of CDR 0.5 cases coincided with increased poly (ADP-ribose) polymerase cleavage but not apoptotic cell injury. In the entorhinal cortex of CDR 5 cases, we found elevation of caspase-1, -2L, -3, -5, -6, -7, -8, and -9 and a greater than 4-fold increase in TUNEL-positive cells. Caspase messenger RNA expression was closely associated with neurofibrillary tangle and, to a lesser extent, neuritic plaque density. CONCLUSIONS: Proapoptotic mechanisms may be at play early in the onset of AD (before overt signs of apoptosis) and may be a conditional factor for later apoptotic cell injury or death. These data have relevance to potential therapeutic interventions for AD using selective caspase inhibitors.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Brain/enzymology , Brain/pathology , Caspases/genetics , Aged , Aged, 80 and over , Apoptosis , Biomarkers , Cognition , Disease Progression , Female , Gene Expression Regulation, Enzymologic , Humans , In Situ Nick-End Labeling , Male , Poly(ADP-ribose) Polymerases/metabolism , RNA, Messenger/analysis
8.
Gene Expr ; 10(5-6): 271-8, 2002.
Article in English | MEDLINE | ID: mdl-12450219

ABSTRACT

Several epidemiologic studies have reported that cyclooxygenase (COX) inhibitors prevent/delay the onset of Alzheimer's disease (AD). Recent experimental studies suggest that these compounds can also diminish amyloid-beta (Abeta) neuropathology in rodent models of AD. To explore the relationship of COX expression to Abeta neuropathology, we crossed mice expressing both mutant amyloid precursor protein [K670N/M671L (APP(swe)] and mutant PS1 (A246E) with mice expressing human COX-2 selectively in neurons. We show here that human COX-2 expression in APP(swe)/PS1/COX-2 mice induces potentiation of brain parenchymal amyloid plaque formation and a greater than twofold increase in prostaglandin E2 production, at 24 months of age. This increased amyloid plaque formation coincided with a preferential elevation of Abeta1-40 and Abeta1-42 with no change in total amyloid precursor protein (APP) expression/content in the brain. Collectively these data suggest that COX-2 influences APP processing and promotes amyloidosis in the brain.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/metabolism , Amyloid/metabolism , Isoenzymes/physiology , Prostaglandin-Endoperoxide Synthases/physiology , Animals , Brain/enzymology , Brain/metabolism , Cyclooxygenase 2 , Dinoprostone/metabolism , Disease Models, Animal , Humans , Image Processing, Computer-Assisted , Inflammation , Isoenzymes/metabolism , Mass Spectrometry , Membrane Proteins , Mice , Mice, Transgenic , Mutation , Prostaglandin-Endoperoxide Synthases/metabolism , RNA, Messenger/metabolism , Time Factors
9.
J Alzheimers Dis ; 34(2): 417-429, 2013.
Article in English | MEDLINE | ID: mdl-23241557

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of death and disability among children and young adults in the United States. In this study, we explored whether changes in the gene expression profile of peripheral blood mononuclear cells (PBMC) may provide a clinically assessable "window" into the brain, reflecting molecular alterations following TBI that might contribute to the onset and progression of TBI clinical complications. We identified three olfactory receptor (OR) TBI biomarkers that are aberrantly down-regulated in PBMC specimens from TBI subjects. Down-regulation of these OR biomarkers in PBMC was correlated with the severity of brain injury and TBI-specific symptoms. A two- biomarker panel comprised of OR11H1 and OR4M1 provided the best criterion for segregating the TBI and control cases with 90% accuracy, 83.3% sensitivity, and 100% specificity. We found that the OR biomarkers are ectopically expressed in multiple brain regions, including the entorhinal-hippocampus system known to play an important role in memory formation and consolidation. Activation of OR4M1 led to attenuation of abnormal tau phosphorylation, possibly through JNK signaling pathway. Our results suggested that addition of the two-OR biomarker model to current diagnostic criteria may lead to improved TBI detection for clinical trials, and decreased expression of OR TBI biomarkers might be associated with TBI-induced tauopathy. Future studies exploring the physiological relevance of OR TBI biomarkers in the normal brain and in the brain following TBI will provide a better understanding of the biological mechanisms underlying TBI and insights into novel therapeutic targets for TBI.


Subject(s)
Brain Injuries/blood , Brain Injuries/epidemiology , Down-Regulation/physiology , Leukocytes, Mononuclear/metabolism , Receptors, Odorant/antagonists & inhibitors , Tauopathies/blood , Tauopathies/epidemiology , Adult , Biomarkers/blood , Brain Injuries/diagnosis , Cohort Studies , Female , Humans , Male , Middle Aged , Receptors, Odorant/biosynthesis , Receptors, Odorant/blood , Tauopathies/diagnosis , Young Adult
10.
Mol Immunol ; 56(4): 619-29, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23911420

ABSTRACT

BACKGROUND: Complement component C5-derived C5a locally generated in the brain has been shown to protect against glutamate-induced neuronal apoptosis and beta-amyloid (Aß) toxicity, but the mechanism is not clear. In this study, we tested the hypothesis that C5a influences upstream signal transduction pathways associated with cAMP-response element-binding protein (CREB) activation, in which alterations of CREB levels are associated with cognitive deterioration in AD. METHODS: CREB signaling pathway, synaptic plasticity and cognitive function were studied in C5a receptor knockout mice (C5aR(-/-)), C5a over expressing mice (C5a/GFAP) and in Tg2576 mice, an AD mouse model. RESULTS: (1) Cognitive function is severely impaired in C5aR(-/-) mice, coincident with the down-regulated CREB/CEBP pathway in brain. (2) Either the application of recombinant-human-C5a (hrC5a) or exogenous expression of C5a in the brain of a mouse model (C5a/GFAP) enhances this pathway. (3) Application of hrC5a in brain slices from Tg2576 mice significantly improves deficits in long-term potentiation (LTP), while this effect is blocked by a specific AMPA receptor antagonist. (4) Searching for a pharmacological approach to locally mediate C5a responses in the brain, we found that low-dose human intravenous immunoglobulin (IVIG) treatment improves synaptic plasticity and cognitive function through C5a-mediated induction of the CREB/CEBP pathway, while the levels of Aß in the brain are not significantly affected. CONCLUSION: This study for the first time provides novel evidence suggesting that C5a may beneficially influence cognitive function in AD through an up-regulation of AMPA-CREB signaling pathway. IVIG may systematically improve cognitive function in AD brain by passing Aß toxicity.


Subject(s)
Alzheimer Disease/therapy , CCAAT-Enhancer-Binding Proteins/metabolism , Complement C5a/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Immunoglobulins, Intravenous/therapeutic use , Receptors, AMPA/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Behavior, Animal/drug effects , Blotting, Western , Brain/drug effects , Brain/metabolism , Brain/pathology , Cells, Cultured , Complement C5a/genetics , Female , Humans , Immunotherapy/methods , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, AMPA/agonists , Receptors, Complement/genetics , Receptors, Complement/metabolism , Signal Transduction/drug effects , Synaptic Potentials/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL