Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Neurotoxicology ; 83: 77-88, 2021 03.
Article in English | MEDLINE | ID: mdl-33417987

ABSTRACT

Low-dose repeated lipopolysaccharide pre-challenge followed by chronic mild stress (LPS/CMS) protocol has been introduced as a rodent model of depression combining the roles of immune activation and chronic psychological stress. However, the impact of this paradigm on cognitive functioning has not been investigated hitherto. METHODS: This study evaluated LPS/CMS-induced cognitive effects and the role of glycogen synthase kinase-3ß (GSK-3ß) activation with subsequent neuroinflammation and pathological tau deposition in the pathogenesis of these effects using lithium (Li) as a tool for GSK-3 inhibition. RESULTS: LPS pre-challenge reduced CMS-induced neuroinflammation, depressive-like behavior and cognitive inflexibility. It also improved spatial learning but increased GSK-3ß expression and exaggerated hyperphosphorylated tau accumulation in hippocampus and prefrontal cortex. Li ameliorated CMS and LPS/CMS-induced depressive and cognitive deficits, reduced GSK-3ß over-expression and tau hyperphosphorylation, impeded neuroinflammation and enhanced neuronal survival. CONCLUSION: This study draws attention to LPS/CMS-triggered cognitive changes and highlights how prior low-dose immune challenge could develop an adaptive capacity to buffer inflammatory damage and maintain the cognitive abilities necessary to withstand threats. This work also underscores the favorable effect of Li (as a GSK-3ß inhibitor) in impeding exaggerated tauopathy and neuroinflammation, rescuing neuronal survival and preserving cognitive functions. Yet, further in-depth studies utilizing different low-dose LPS challenge schedules are needed to elucidate the complex interactions between immune activation and chronic stress exposure.


Subject(s)
Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Cognition/drug effects , Cognitive Dysfunction/prevention & control , Depression/prevention & control , Encephalitis/prevention & control , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Hippocampus/drug effects , Lithium Chloride/pharmacology , Protein Kinase Inhibitors/pharmacology , Tauopathies/prevention & control , Animals , Cerebral Cortex/enzymology , Cerebral Cortex/physiopathology , Chronic Disease , Cognitive Dysfunction/enzymology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/physiopathology , Depression/enzymology , Depression/etiology , Depression/physiopathology , Disease Models, Animal , Encephalitis/enzymology , Encephalitis/etiology , Encephalitis/physiopathology , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/enzymology , Hippocampus/physiopathology , Inflammation Mediators/metabolism , Lipopolysaccharides , Male , Phosphorylation , Rats, Wistar , Spatial Learning/drug effects , Stress, Psychological/complications , Stress, Psychological/psychology , Tauopathies/enzymology , Tauopathies/etiology , Tauopathies/physiopathology , tau Proteins/metabolism
2.
Aging (Albany NY) ; 11(21): 9672-9688, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31714894

ABSTRACT

Protein phosphatase 2A (PP2A) is the major tau phosphatase. Its activity toward tau is regulated by the methylation of PP2A catalytic subunit (PP2Ac) at Leu309. Protein phosphatase methylesterase-1 (PME-1) demethylates PP2Ac and suppresses its activity. We previously found that glycogen synthase kinase-3ß (GSK-3ß) suppresses PME-1 expression. However, the underlying molecular mechanism is unknown. In the present study, we analyzed the promoter of PME-1 gene and found that human PME-1 promoter contains two lymphoid enhancer binding factor-1/T-cell factor (LEF1/TCF) cis-elements in which ß-catenin serves as a co-activator. ß-catenin acted on these two cis-elements and promoted PME-1 expression. GSK-3ß phosphorylated ß-catenin and suppressed its function in promoting PME-1 expression. Inhibition and activation of GSK-3ß by PI3K-AKT pathway promoted and suppressed, respectively, PME-1 expression in primary cultured neurons, SH-SY5Y cells and in the mouse brain. These findings suggest that GSK-3ß phosphorylates ß-catenin and suppresses its function on PME-1 expression, resulting in an increase of PP2Ac methylation.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Tauopathies/etiology , beta Catenin/metabolism , Animals , Base Sequence , Carboxylic Ester Hydrolases/genetics , HEK293 Cells , Humans , Mice, Transgenic , Phosphatidylinositol 3-Kinases/metabolism , Primary Cell Culture , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Tauopathies/enzymology
3.
Alzheimers Res Ther ; 11(1): 58, 2019 06 29.
Article in English | MEDLINE | ID: mdl-31253191

ABSTRACT

BACKGROUND: Tau stabilizes microtubules; however, in Alzheimer's disease (AD) and tauopathies, tau becomes hyperphosphorylated, aggregates, and results in neuronal death. Our group recently uncovered a unique interaction between polyamine metabolism and tau fate. Polyamines exert an array of physiological effects that support neuronal function and cognitive processing. Specific stimuli can elicit a polyamine stress response (PSR), resulting in altered central polyamine homeostasis. Evidence suggests that elevations in polyamines following a short-term stressor are beneficial; however, persistent stress and subsequent PSR activation may lead to maladaptive polyamine dysregulation, which is observed in AD, and may contribute to neuropathology and disease progression. METHODS: Male and female mice harboring tau P301L mutation (rTg4510) were examined for a tau-induced central polyamine stress response (tau-PSR). The direct effect of tau-PSR byproducts on tau fibrillization and oligomerization were measured using a thioflavin T assay and a N2a split superfolder GFP-Tau (N2a-ssGT) cell line, respectively. To therapeutically target the tau-PSR, we bilaterally injected caspase 3-cleaved tau truncated at aspartate 421 (AAV9 Tau ΔD421) into the hippocampus and cortex of spermidine/spermine-N1-acetyltransferase (SSAT), a key regulator of the tau-PSR, knock out (SSAT-/-), and wild type littermates, and the effects on tau neuropathology, polyamine dysregulation, and behavior were measured. Lastly, cellular models were employed to further examine how SSAT repression impacted tau biology. RESULTS: Tau induced a unique tau-PSR signature in rTg4510 mice, notably in the accumulation of acetylated spermidine. In vitro, higher-order polyamines prevented tau fibrillization but acetylated spermidine failed to mimic this effect and even promoted fibrillization and oligomerization. AAV9 Tau ΔD421 also elicited a unique tau-PSR in vivo, and targeted disruption of SSAT prevented the accumulation of acetylated polyamines and impacted several tau phospho-epitopes. Interestingly, SSAT knockout mice presented with altered behavior in the rotarod task, the elevated plus maze, and marble burying task, thus highlighting the impact of polyamine homeostasis within the brain. CONCLUSION: These data represent a novel paradigm linking tau pathology and polyamine dysfunction and that targeting specific arms within the polyamine pathway may serve as new targets to mitigate certain components of the tau phenotype.


Subject(s)
Acetyltransferases/metabolism , Polyamines/metabolism , Stress, Physiological , Tauopathies/enzymology , Acetyltransferases/genetics , Animals , Female , Hippocampus/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Protein Aggregation, Pathological/metabolism , tau Proteins/metabolism
4.
Acta Neuropathol Commun ; 6(1): 103, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30270813

ABSTRACT

Chemotherapy-induced cognitive impairment (CICI) is a commonly reported neurotoxic side effect of chemotherapy, occurring in up to 75% cancer patients. CICI manifests as decrements in working memory, executive functioning, attention, and processing speed, and greatly interferes with patients' daily performance and quality of life. Currently no treatment for CICI has been approved by the US Food and Drug Administration. We show here that treatment with a brain-penetrating histone deacetylase 6 (HDAC6) inhibitor for two weeks was sufficient to fully reverse cisplatin-induced cognitive impairments in male mice, as demonstrated in the Y-maze test of spontaneous alternation, the novel object/place recognition test, and the puzzle box test. Normalization of cognitive impairment was associated with reversal of cisplatin-induced synaptosomal mitochondrial deficits and restoration of synaptic integrity. Mechanistically, cisplatin induced deacetylation of the microtubule protein α-tubulin and hyperphosphorylation of the microtubule-associated protein tau. These cisplatin-induced changes were reversed by HDAC6 inhibition. Our data suggest that inhibition of HDAC6 restores microtubule stability and reverses tau phosphorylation, leading to normalization of synaptosomal mitochondrial function and synaptic integrity and thereby to reversal of CICI. Remarkably, our results indicate that short-term daily treatment with the HDAC6 inhibitor was sufficient to achieve prolonged reversal of established behavioral, structural and functional deficits induced by cisplatin. Because the beneficial effects of HDAC6 inhibitors as add-ons to cancer treatment have been demonstrated in clinical trials, selective targeting of HDAC6 with brain-penetrating inhibitors appears a promising therapeutic approach for reversing chemotherapy-induced neurotoxicity while enhancing tumor control.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/toxicity , Cognitive Dysfunction , Enzyme Inhibitors/therapeutic use , Histone Deacetylase 6/metabolism , Tauopathies/enzymology , Animals , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/enzymology , Disease Models, Animal , Disks Large Homolog 4 Protein/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/blood , Green Fluorescent Proteins/metabolism , Histone Deacetylase 6/ultrastructure , Hydroxamic Acids/blood , Hydroxamic Acids/therapeutic use , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Pyrimidines/blood , Pyrimidines/therapeutic use , Recombinant Fusion Proteins/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism , Synaptosomes/pathology , Synaptosomes/ultrastructure , Tauopathies/chemically induced , Tauopathies/drug therapy , Time Factors , Tubulin/metabolism , tau Proteins/metabolism
5.
J Alzheimers Dis ; 64(s1): S481-S489, 2018.
Article in English | MEDLINE | ID: mdl-29758943

ABSTRACT

Tauopathies belong to a large group of neurodegenerative diseases characterized by progressive accumulation of hyperphosphorylated tau. Tau is a microtubule binding protein which is necessary for their assembly and stability. However, tau affinity for microtubules mainly depends on its phosphorylation status, which is the result of a delicate balance between kinases and phosphatases activity. Any significant changes in this equilibrium can promote tau fibrillation, aggregation, neuronal dysfunction, and ultimately neuronal loss. Despite intensive research, the molecular mechanism(s) leading to tau hyperphosphorylation are still unknown and there is no cure for these diseases. Development of an effective strategy that successfully prevents tau excessive phosphorylation and/or tau aggregation may offer a real therapeutic opportunity for these less investigated neurodegenerative conditions. Beside tau, chronic brain inflammation is a common feature of all tauopathies and 5-lipoxygenase, an inflammatory enzyme, is upregulated in brain regions affected by tau pathology. Recently, in vitro studies and preclinical investigations with animal models of tauopathy have implicated 5-lipoxygenase in the regulation of tau phosphorylation through activation of the cyclin-dependent kinase 5 pathway, supporting the novel hypothesis that this protein is a promising therapeutic target for the treatment of tauopathies. In this article, we will discuss the contribution of the 5-lipoxygenase signaling pathway in the development of tau neuropathology, and the promising potential that drugs targeting this enzyme activation hold as a novel disease-modifying therapeutic approach for tauopathies.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Tauopathies/enzymology , Animals , Humans , Tauopathies/drug therapy , tau Proteins/metabolism
6.
Biomol Concepts ; 9(1): 1-11, 2018 Mar 05.
Article in English | MEDLINE | ID: mdl-29509544

ABSTRACT

The microtubule-associated protein Tau plays a crucial role in stabilizing neuronal microtubules. In Tauopathies, Tau loses its ability to bind microtubules, detach from them and forms intracellular aggregates. Increasing evidence in recent years supports the notion that Tau pathology spreading throughout the brain in AD and other Tauopathies is the consequence of the propagation of specific Tau species along neuroanatomically connected brain regions in a so-called "prion-like" manner. A number of steps are assumed to be involved in this process, including secretion, cellular uptake, transcellular transfer and/or seeding, although the precise mechanisms underlying propagation of Tau pathology are not fully understood yet. This review summarizes recent evidence on the nature of the specific Tau species that are propagated and the different mechanisms of Tau pathology spreading.


Subject(s)
tau Proteins/metabolism , Animals , Brain/pathology , Cells, Cultured , Humans , Mice , Protein Aggregation, Pathological , Tauopathies/enzymology , Tauopathies/pathology
7.
Mol Neurobiol ; 55(7): 5926-5936, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29128902

ABSTRACT

Brain accumulation of increasing amount of phosphorylated microtubule associated tau protein is one the major hallmark lesions of Alzheimer's disease (AD) and related tauopathies. Consistent evidence from clinical and animal studies has shown that neuroinflammation characterizes these diseases. The 5-lipoxygenase (5LO) is an enzyme protein whose metabolic products are lipids with potent inflammatory actions. Previously, we showed that blockade of 5LO activation ameliorates the phenotype of the htau transgenic mice. Here, by employing a vector system to overexpress 5LO in the brain of the same mouse model, we investigated its role and contribution to their behavioral deficits and development of tau neuropathology. Compared with controls, 5LO gene targeted mice manifested significant impairments in their memory and learning ability. On the other hand, brain tissues of the same mice had higher 5LO protein level and activity which resulted in intense neuroinflammation and synaptic pathology. Further, the same mice had a significant elevation of tau phosphorylation, which associated with the activation of the cdk5 kinase and an accumulation of insoluble tau. The functional involvement of this kinase in the 5LO-dependent tau phosphorylation was confirmed in neuronal cells. Taken together, our findings demonstrate that neuronal 5LO is directly involved in tau phosphorylation and tau neuropathology, and for this reason, it should be considered a good therapeutic target for tauopathies.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Tauopathies/enzymology , Tauopathies/pathology , Animals , Behavior, Animal , Brain/pathology , Cyclin-Dependent Kinase 5/metabolism , Disease Models, Animal , Humans , Inflammation/pathology , Mice, Transgenic , Phenotype , Phosphorylation , Signal Transduction , Synapses/metabolism
8.
J Neuroinflammation ; 14(1): 118, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28599675

ABSTRACT

BACKGROUND: The innate immune system is known to be involved early in the pathogenesis of Alzheimer's disease (AD) and other neurodegenerative disorders. The inflammatory response in the central nervous system can be measured postmortem or through a series of inflammatory mediator surrogates. YKL-40 (also named Chitinase-3-like I) has been frequently investigated in body fluids as a surrogate marker of neuroinflammation in AD and other neurological disorders. However, the expression pattern of YKL-40 in the human brain with neurodegenerative pathology remains poorly investigated. Our aim was to study the cellular expression pattern of YKL-40 in the brain of patients with clinical and neuropathological criteria for AD (n = 11); three non-AD tauopathies: Pick's disease (PiD; n = 8), corticobasal degeneration (CBD; n = 8) and progressive supranuclear palsy (PSP; n = 9) and a group of neurologically healthy controls (n = 6). METHODS: Semiquantitative neuropathological evaluation and quantitative confocal triple immunofluorescence studies were performed. An in-house algorithm was used to detect and quantify pathology burden of random regions of interest on a full tissue-section scan. Kruskal-Wallis and Dunn's multiple comparison tests were performed for colocalization and quantification analyses. RESULTS: We found that brain YKL-40 immunoreactivity was observed in a subset of astrocytes in all four diseases and in controls. There was a strong colocalization between YKL-40 and the astroglial marker GFAP but not with neuronal nor microglial markers. Intriguingly, YKL-40-positive astrocytes were tau-negative in PSP, CBD and PiD. The number of YKL-40-positive astrocytes was increased in tauopathies compared with that in controls. A positive correlation was found between YKL-40 and tau immunoreactivities. CONCLUSIONS: This study confirms that YKL-40 is expressed by a subset of astrocytes in AD and other tauopathies. YKL-40 expression is elevated in several neurodegenerative conditions and correlates with tau pathology.


Subject(s)
Alzheimer Disease/enzymology , Astrocytes/enzymology , Brain/enzymology , Chitinase-3-Like Protein 1/biosynthesis , Gene Expression Regulation, Enzymologic , Tauopathies/enzymology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Astrocytes/pathology , Brain/pathology , Chitinase-3-Like Protein 1/genetics , Humans , Tauopathies/genetics , Tauopathies/pathology
9.
J Neurosci ; 35(44): 14842-60, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26538654

ABSTRACT

Tau accumulation remains one of the closest correlates of neuronal loss in Alzheimer's disease. In addition, tau associates with several other neurodegenerative diseases, collectively known as tauopathies, in which clinical phenotypes manifest as cognitive impairment, behavioral disturbances, and motor impairment. Polyamines act as bivalent regulators of cellular function and are involved in numerous biological processes. The regulation of the polyamines system can become dysfunctional during disease states. Arginase 1 (Arg1) and nitric oxide synthases compete for l-arginine to produce either polyamines or nitric oxide, respectively. Herein, we show that overexpression of Arg1 using adeno-associated virus (AAV) in the CNS of rTg4510 tau transgenic mice significantly reduced phospho-tau species and tangle pathology. Sustained Arg1 overexpression decreased several kinases capable of phosphorylating tau, decreased inflammation, and modulated changes in the mammalian target of rapamycin and related proteins, suggesting activation of autophagy. Arg1 overexpression also mitigated hippocampal atrophy in tau transgenic mice. Conversely, conditional deletion of Arg1 in myeloid cells resulted in increased tau accumulation relative to Arg1-sufficient mice after transduction with a recombinant AAV-tau construct. These data suggest that Arg1 and the polyamine pathway may offer novel therapeutic targets for tauopathies.


Subject(s)
Arginase/biosynthesis , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Tauopathies/enzymology , Tauopathies/pathology , tau Proteins/metabolism , Animals , Arginase/genetics , HeLa Cells , Hippocampus/enzymology , Hippocampus/pathology , Humans , Mice , Mice, Transgenic , Tauopathies/genetics , tau Proteins/genetics
11.
J Med Chem ; 58(11): 4590-609, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25923950

ABSTRACT

Fyn is a member of the Src-family of nonreceptor protein-tyrosine kinases. Its abnormal activity has been shown to be related to various human cancers as well as to severe pathologies, such as Alzheimer's and Parkinson's diseases. Herein, a structure-based drug design protocol was employed aimed at identifying novel Fyn inhibitors. Two hits from commercial sources (1, 2) were found active against Fyn with K(i) of about 2 µM, while derivative 4a, derived from our internal library, showed a K(i) of 0.9 µM. A hit-to-lead optimization effort was then initiated on derivative 4a to improve its potency. Slightly modifications rapidly determine an increase in the binding affinity, with the best inhibitors 4c and 4d having K(i)s of 70 and 95 nM, respectively. Both compounds were found able to inhibit the phosphorylation of the protein Tau in an Alzheimer's model cell line and showed antiproliferative activities against different cancer cell lines.


Subject(s)
Adenosine Triphosphate/metabolism , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Tauopathies/drug therapy , Antineoplastic Agents/chemistry , Binding Sites , Cell Proliferation/drug effects , Humans , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Neoplasms/enzymology , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-fyn/metabolism , Pyrazoles/chemistry , Pyrimidines/chemistry , Signal Transduction/drug effects , Structure-Activity Relationship , Tauopathies/enzymology , Tumor Cells, Cultured
12.
Biol Psychiatry ; 78(10): 693-701, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-25802082

ABSTRACT

BACKGROUND: 5-Lipoxygenase (5-LO) is a protein widely distributed in the central nervous system where it modulates amyloidosis and memory impairments in transgenic mouse models of Alzheimer's disease. However, no data are available as to whether 5-LO is elevated in human tauopathy or if it directly influences tau pathology in a relevant model of the disease. METHODS: We assayed 5-LO levels in brain samples from patients with tauopathy and transgenic tau mice, and we evaluated the effect of 5-LO pharmacologic inhibition on the phenotype of these mice. RESULTS: The 5-LO protein is upregulated in human tauopathy and transgenic tau mice brains. Pharmacologic blockade of 5-LO in tau mice resulted in significant memory improvement, rescue of synaptic integrity and dysfunction, and reduction of tau pathology via a cdk5-dependent mechanism. CONCLUSIONS: These results establish a key role of 5-LO in the development of the tau pathology phenotype and demonstrate it to be a novel viable therapeutic target for the pharmacologic treatment of human tauopathy.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Brain/enzymology , Memory/physiology , Neurons/enzymology , Tauopathies/enzymology , tau Proteins/metabolism , Animals , Brain/drug effects , Brain/metabolism , Cerebellum/drug effects , Cerebellum/enzymology , Cerebellum/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Cyclin-Dependent Kinase 5/metabolism , Disease Models, Animal , Encephalitis/enzymology , Excitatory Postsynaptic Potentials/drug effects , Humans , Hydroxyurea/analogs & derivatives , Hydroxyurea/pharmacology , Lipoxygenase Inhibitors/pharmacology , Memory/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Phenotype , Phosphorylation/drug effects , Synapses/drug effects , Synapses/metabolism , Synapses/pathology , Tauopathies/metabolism , tau Proteins/genetics
13.
Exp Neurol ; 265: 48-58, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25483398

ABSTRACT

Alzheimer's disease is the most prevalent tauopathy and cause of dementia. We investigate the hypothesis that reactivation of plasticity can restore function in the presence of neuronal damage resulting from tauopathy. We investigated two models with tau hyperphosphorylation, aggregation and neurodegeneration: a transgenic mouse model in which the mutant P301S tau is expressed in neurons (Tg P301S), and a model in which an adeno-associated virus expressing P301S tau (AAV-P301S) was injected in the perirhinal cortex, a region critical for object recognition (OR) memory. Both models show profound loss of OR memory despite only 15% neuronal loss in the Tg P301S and 26% in AAV-P301S-injected mice. Recordings from perirhinal cortex slices of 3month-old P301S transgenic mice showed a diminution in synaptic transmission following temporal stimulation. Chondroitinase ABC (ChABC) can reactivate plasticity and affect memory through actions on perineuronal nets. ChABC was injected into the perirhinal cortex and animals were tested for OR memory 1week later, demonstrating restoration of OR memory to normal levels. Synaptic transmission indicated by fEPSP amplitude was restored to control levels following ChABC treatment. ChABC did not affect the progression of neurodegenerative tauopathy. These findings suggest that increasing plasticity by manipulation of perineuronal nets offers a novel therapeutic approach to the treatment of memory loss in neurodegenerative disorders.


Subject(s)
Cerebral Cortex/enzymology , Chondroitin ABC Lyase/administration & dosage , Memory/physiology , Nerve Net/enzymology , Neuronal Plasticity/physiology , Tauopathies/enzymology , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Humans , Injections, Intraventricular , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/drug effects , Nerve Net/pathology , Neuronal Plasticity/drug effects , Organ Culture Techniques , Tauopathies/drug therapy , Tauopathies/pathology
14.
Neurobiol Dis ; 67: 119-32, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24704314

ABSTRACT

Tau.P301L transgenic mice suffer precocious mortality between ages 8 and 11 months, resulting from upper airway defects caused by tauopathy in autonomic brainstem circuits that control breathing (Dutschmann et al., 2010). In individual mice, the clinical phenotype evolves progressively and rapidly (3-6weeks) from clasping, over general motor impairment to severe reduction in body-weight into the terminal phase that announces imminent death (<3days). Surprisingly, co-expression of GSK3ß with Tau.P301L significantly prolonged survival of bigenic biGT mice (Terwel et al., 2008), which we here assign to delayed development of brainstem tauopathy. Eventually, brainstem tauopathy became as prominent in old biGT mice in the specified brainstem nuclei as in the parental Tau.P301L mice, resulting in similar clinical deterioration and terminal phase preceding death, although at later age. Biochemically, in both genotypes the pathway to neurofibrillary tangles and neuropil threads was similar: phosphorylation of protein Tau and formation of soluble oligomers and insoluble aggregates, ending in the typical tangles and threads of tauopathy. The extra GSK3ß activity led to expected increased phosphorylation of protein Tau, particularly at residues S262 and S396, which we must conclude to delay the aggregation of protein Tau in the brainstem of aging biGT mice. The unexpected, paradoxical alleviation of the brainstem problems in biGT mice allowed them to grow older and thereby develop more severe tauopathy in forebrain than Tau.P301L mice, which succumb at younger age.


Subject(s)
Brain Stem/enzymology , Glycogen Synthase Kinase 3/metabolism , Tauopathies/enzymology , tau Proteins/chemistry , tau Proteins/metabolism , Animals , Brain/enzymology , Brain/metabolism , Brain Stem/metabolism , Female , Glycogen Synthase Kinase 3 beta , Male , Mice , Mice, Transgenic , Phosphorylation , Survival Analysis , Tauopathies/metabolism
15.
Aging Cell ; 13(1): 49-59, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23919677

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by pathological deposits of ß-amyloid (Aß) in senile plaques, intracellular neurofibrillary tangles (NFTs) comprising hyperphosphorylated aggregated tau, synaptic dysfunction and neuronal death. Substantial evidence indicates that disrupted neuronal calcium homeostasis is an early event in AD that could mediate synaptic dysfunction and neuronal toxicity. Sodium calcium exchangers (NCXs) play important roles in regulating intracellular calcium, and accumulating data suggests that reduced NCX function, following aberrant proteolytic cleavage of these exchangers, may contribute to neurodegeneration. Here, we show that elevated calpain, but not caspase-3, activity is a prominent feature of AD brain. In addition, we observe increased calpain-mediated cleavage of NCX3, but not a related family member NCX1, in AD brain relative to unaffected tissue and that from other neurodegenerative conditions. Moreover, the extent of NCX3 proteolysis correlated significantly with amounts of Aß1-42. We also show that exposure of primary cortical neurons to oligomeric Aß1-42 results in calpain-dependent cleavage of NCX3, and we demonstrate that loss of NCX3 function is associated with Aß toxicity. Our findings suggest that Aß mediates calpain cleavage of NCX3 in AD brain and therefore that reduced NCX3 activity could contribute to the sustained increases in intraneuronal calcium concentrations that are associated with synaptic and neuronal dysfunction in AD.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Calpain/metabolism , Sodium-Calcium Exchanger/metabolism , Aged , Aged, 80 and over , Amyloid beta-Peptides/toxicity , Animals , Brain/drug effects , Brain/enzymology , Brain/pathology , Calcium-Binding Proteins/metabolism , Caspase 3/metabolism , Cells, Cultured , Female , Gene Knockdown Techniques , Humans , Male , Middle Aged , Oligonucleotides, Antisense/pharmacology , Postmortem Changes , Protein Subunits/metabolism , Rats , Spectrin/metabolism , Substrate Specificity/drug effects , Tauopathies/enzymology , Tauopathies/pathology
16.
J Biol Chem ; 287(42): 35444-35453, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22918830

ABSTRACT

Both neuroprotective and neurotoxic roles have previously been described for histone deacetylase-1 (HDAC1). Here we report that HDAC1 expression is elevated in vulnerable brain regions of two mouse models of neurodegeneration, the R6/2 model of Huntington disease and the Ca(2+)/calmodulin-dependent protein kinase (CaMK)/p25 double-transgenic model of tauopathic degeneration, suggesting a role in promoting neuronal death. Indeed, elevating HDAC1 expression by ectopic expression promotes the death of otherwise healthy cerebellar granule neurons and cortical neurons in culture. The neurotoxic effect of HDAC1 requires interaction and cooperation with HDAC3, which has previously been shown to selectively induce the death of neurons. HDAC1-HDAC3 interaction is greatly elevated under conditions of neurodegeneration both in vitro and in vivo. Furthermore, the knockdown of HDAC3 suppresses HDAC1-induced neurotoxicity, and the knockdown of HDAC1 suppresses HDAC3 neurotoxicity. As described previously for HDAC3, the neurotoxic effect of HDAC1 is inhibited by treatment with IGF-1, the expression of Akt, or the inhibition of glycogen synthase kinase 3ß (GSK3ß). In addition to HDAC3, HDAC1 has been shown to interact with histone deacetylase-related protein (HDRP), a truncated form of HDAC9, whose expression is down-regulated during neuronal death. In contrast to HDAC3, the interaction between HDRP and HDAC1 protects neurons from death, an effect involving acquisition of the deacetylase activity of HDAC1 by HDRP. We find that elevated HDRP inhibits HDAC1-HDAC3 interaction and prevents the neurotoxic effect of either of these two proteins. Together, our results suggest that HDAC1 is a molecular switch between neuronal survival and death. Its interaction with HDRP promotes neuronal survival, whereas interaction with HDAC3 results in neuronal death.


Subject(s)
Cell Cycle , Cerebellar Cortex/enzymology , Histone Deacetylase 1/metabolism , Nerve Tissue Proteins/metabolism , Neurons/enzymology , Tauopathies/enzymology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Death/genetics , Cerebellar Cortex/pathology , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Histone Deacetylase 1/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Tauopathies/genetics , Tauopathies/pathology
17.
J Biol Chem ; 287(25): 20931-41, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22535953

ABSTRACT

Protective proteases are key elements of protein quality control pathways that are up-regulated, for example, under various protein folding stresses. These proteases are employed to prevent the accumulation and aggregation of misfolded proteins that can impose severe damage to cells. The high temperature requirement A (HtrA) family of serine proteases has evolved to perform important aspects of ATP-independent protein quality control. So far, however, no HtrA protease is known that degrades protein aggregates. We show here that human HTRA1 degrades aggregated and fibrillar tau, a protein that is critically involved in various neurological disorders. Neuronal cells and patient brains accumulate less tau, neurofibrillary tangles, and neuritic plaques, respectively, when HTRA1 is expressed at elevated levels. Furthermore, HTRA1 mRNA and HTRA1 activity are up-regulated in response to elevated tau concentrations. These data suggest that HTRA1 is performing regulated proteolysis during protein quality control, the implications of which are discussed.


Subject(s)
Nerve Tissue Proteins/chemistry , Protein Folding , Proteolysis , Serine Endopeptidases/chemistry , tau Proteins/chemistry , Brain/metabolism , Brain/pathology , Gene Expression Regulation, Enzymologic , High-Temperature Requirement A Serine Peptidase 1 , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurites/enzymology , Neurites/pathology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Tauopathies/enzymology , Tauopathies/pathology , tau Proteins/genetics , tau Proteins/metabolism
18.
Prog Mol Biol Transl Sci ; 106: 343-79, 2012.
Article in English | MEDLINE | ID: mdl-22340724

ABSTRACT

Alzheimer's Disease (AD) is characterized by progressive loss of cognitive function, linked to marked neuronal loss. Pathological hallmarks of the disease are the accumulation of the amyloid-ß (Aß) peptide in the form of amyloid plaques and the intracellular formation of neurofibrillary tangles (NFTs). Accumulating evidence supports a key role for protein phosphorylation in both the normal and pathological actions of Aß as well as the formation of NFTs. NFTs contain hyperphosphorylated forms of the microtubule-binding protein tau, and phosphorylation of tau by several different kinases leads to its aggregation. The protein kinases involved in the generation and/or actions of tau or Aß are viable drug targets to prevent or alleviate AD pathology. However, it has also been recognized that the protein phosphatases that reverse the actions of these protein kinases are equally important. Here, we review recent advances in our understanding of serine/threonine and tyrosine protein phosphatases in the pathology of AD.


Subject(s)
Alzheimer Disease/enzymology , Nerve Tissue Proteins/physiology , Phosphoprotein Phosphatases/physiology , Protein Processing, Post-Translational , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Calcineurin/physiology , Calcineurin Inhibitors , Disease Models, Animal , Dopaminergic Neurons/enzymology , Dopaminergic Neurons/physiology , Enzyme Inhibitors/therapeutic use , Humans , Mice , Mice, Transgenic , Nuclear Proteins/physiology , Phosphoproteins/metabolism , Phosphorylation , Protein Phosphatase 1/physiology , Protein Phosphatase 2/physiology , Protein Tyrosine Phosphatases, Non-Receptor/physiology , Tauopathies/enzymology , tau Proteins/metabolism
19.
J Neuropathol Exp Neurol ; 71(2): 116-29, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22249463

ABSTRACT

Traumatic brain injury (TBI) is a major environmental risk factor for subsequent development of Alzheimer disease (AD). Pathological features that are common to AD and many tauopathies are neurofibrillary tangles (NFTs) and neuropil threads composed of hyperphosphorylated tau. Axonal accumulations of total and phospho-tau have been observed within hours to weeks, and intracytoplasmic NFTs have been documented years after severe TBI in humans. We previously reported that controlled cortical impact TBI accelerated tau pathology in young 3xTg-AD mice. Here, we used this TBI mouse model to investigate mechanisms responsible for increased tau phosphorylation and accumulation after brain trauma. We found that TBI resulted in abnormal axonal accumulation of several kinases that phosphorylate tau. Notably, c-Jun N-terminal kinase (JNK) was markedly activated in injured axons and colocalized with phospho-tau. We found that moderate reduction of JNK activity (40%) by a peptide inhibitor, D-JNKi1, was sufficient to reduce total and phospho-tau accumulations in axons of these mice with TBI. Longer-term studies will be required to determine whether reducing acute tau pathology proves beneficial in brain trauma.


Subject(s)
Brain Injuries/complications , Enzyme Inhibitors/therapeutic use , JNK Mitogen-Activated Protein Kinases/metabolism , Peptides/therapeutic use , Tauopathies , Amyloid beta-Peptides/genetics , Animals , Brain Injuries/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Disability Evaluation , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Peptides/pharmacology , Phosphorylation/drug effects , Presenilin-1/genetics , Severity of Illness Index , Tauopathies/drug therapy , Tauopathies/enzymology , Tauopathies/etiology , tau Proteins/genetics , tau Proteins/metabolism
20.
Neurobiol Aging ; 33(3): 624.e23-34, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21530001

ABSTRACT

Pathological hyperphosphorylation and aggregation of the tau protein is associated with dementia and can be the central cause of neurodegeneration. Here, we examined potential alterations in the level of the cholinergic enzyme acetylcholinesterase (AChE) in the brain of transgenic mice (Tg-VLW) expressing human tau mutations. Overexpression of mutant hyperphosphorylated tau (P-tau) led to an increase in the activity of AChE in the brain of Tg-VLW mice, paralleled by an increase in AChE protein and transcripts; whereas the levels of the enzyme choline acetyltransferase remained unaffected. VLW tau overexpression in SH-SY5Y cells also increased AChE activity levels. All major molecular forms of AChE were increased in the Tg-VLW mice, including tetrameric AChE, which is the major species involved in hydrolysis of acetylcholine in the brain. Colocalization of human P-tau and AChE supports the conclusion that P-tau can act to increase AChE. This study is the first direct evidence of a modulatory effect of P-tau on brain AChE expression.


Subject(s)
Acetylcholinesterase/genetics , Gene Expression Regulation, Enzymologic , Tauopathies/genetics , Tauopathies/metabolism , tau Proteins/genetics , Acetylcholinesterase/biosynthesis , Animals , Cell Line, Tumor , Disease Models, Animal , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Gene Expression Regulation, Enzymologic/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroblastoma/enzymology , Neuroblastoma/genetics , Neuroblastoma/metabolism , Tauopathies/enzymology , tau Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...