Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Alzheimers Dement (Amst) ; 11: 53-60, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30623019

ABSTRACT

INTRODUCTION: Total tau (t-tau) and phosphorylated tau (p-tau) are abnormally elevated in the brain and cerebrospinal fluid of individuals with Alzheimer's disease (AD). Tau is also present in the salivary gland tissue and saliva, and salivary measures might produce an accurate, accessible, and inexpensive biomarker. METHODS: Using unstimulated saliva and Western blot analysis, we quantified the p-tau/t-tau ratio at different phosphorylation sites. RESULTS: We found that for one phosphorylation site, S396, p-tau/t-tau ratio was significantly elevated in patients with AD compared with normal elderly control subjects. The elevation in saliva, however, did not correlate with cerebrospinal fluid tau or with brain measures such as hippocampal volume. DISCUSSION: There is significant elevation of p-tau/t-tau ratio for the S396 phosphorylation site. Large variation in the AD salivary tau levels, however, limits the utility of this test as a clinical biomarker.

2.
Mol Cell Neurosci ; 89: 1-8, 2018 06.
Article in English | MEDLINE | ID: mdl-29577984

ABSTRACT

In Alzheimer's disease (AD) tau protein hyperphosphorylation causes neurofibrillary tangle formation, microtubule instability and neurodegeneration. Determining the mechanism of tau hyperphosphorylation will provide a better understanding of AD pathology. Cystatin C (CysC) is a risk factor for late-onset AD and its level is upregulated in the brains of AD patients. The role of CysC is AD pathogenesis is not known. In this study, we found that CysC level is upregulated in 3xTg-AD mouse brain. We demonstrate that CysC does not affect cellular Aß production. However, when overexpressed in neuron (NGF-differentiated PC12 cells), CysC inhibits turnover of GSK3ß, promotes GSK3ß-catalyzed tau phosphorylation at Ser396/404 and causes microtubule instability. Our data provide a novel insight into the role of CysC in AD pathogenesis.


Subject(s)
Alzheimer Disease/metabolism , Cystatin C/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Microtubules/metabolism , Neurons/metabolism , Protein Processing, Post-Translational , tau Proteins/metabolism , Animals , HEK293 Cells , Humans , Mice , Microtubules/drug effects , Neurons/drug effects , PC12 Cells , Phosphorylation , Proteolysis , Rats
3.
Front Aging Neurosci ; 9: 258, 2017.
Article in English | MEDLINE | ID: mdl-28824419

ABSTRACT

Synaptic transmission requires intricate coordination of the components involved in processing of incoming signals, formation and stabilization of synaptic machinery, neurotransmission and in all related signaling pathways. Changes to any of these components cause synaptic imbalance and disruption of neuronal circuitry. Extensive studies at the neuromuscular junction (NMJ) have greatly aided in the current understanding of synapses and served to elucidate the underlying physiology as well as associated adaptive and homeostatic processes. The heparan sulfate proteoglycan agrin is a vital component of the NMJ, mediating synaptic formation and maintenance in both brain and muscle, but very little is known about direct control of its expression. Here, we investigated the relationship between agrin and transcription factor early growth response-1 (Egr-1), as Egr-1 regulates the expression of many genes involved in synaptic homeostasis and plasticity. Using chromatin immunoprecipitation (ChIP), cell culture with cell lines derived from brain and muscle, and animal models, we show that Egr-1 binds to the AGRN gene locus and suppresses its expression. When compared with wild type (WT), mice deficient in Egr-1 (Egr-1-/-) display a marked increase in AGRN mRNA and agrin full-length and cleavage fragment protein levels, including the 22 kDa, C-terminal fragment in brain and muscle tissue homogenate. Because agrin is a crucial component of the NMJ, we explored possible physiological implications of the Egr-1-agrin relationship. In the diaphragm, Egr-1-/- mice display increased NMJ motor endplate density, individual area and area of innervation. In addition to increased density, soleus NMJs also display an increase in fragmented and faint endplates in Egr-1-/- vs. WT mice. Moreover, the soleus NMJ electrophysiology of Egr-1-/- mice revealed increased quantal content and motor testing showed decreased movement and limb muscle strength compared with WT. This study provides evidence for the potential involvement of a novel Egr-1-agrin pathway in synaptic homeostatic and compensatory mechanisms at the NMJ. Synaptic homeostasis is greatly affected by the process of aging. These and other data suggest that changes in Egr-1 expression may directly or indirectly promote age-related pathologies.

4.
Oncotarget ; 8(30): 48533, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28562319
5.
Am J Pathol ; 187(8): 1828-1847, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28641077

ABSTRACT

A sporadic form of Alzheimer disease (AD) and vascular dementia share many risk factors, and their pathogenic mechanisms are suggested to be related. Transcription factor early growth response 1 (Egr-1) regulates various vascular pathologies and is up-regulated in both AD brains and AD mouse models; however, its role in AD pathogenesis is unclear. Herein, we report that silencing of Egr-1 in the hippocampus by shRNA reduces tau phosphorylation, lowers amyloid-ß (Aß) pathology, and improves cognition in the 3xTg-AD mouse model. Egr-1 silencing does not affect levels of cyclin-dependent protein kinase 5 (Cdk5), glycogen synthase kinase 3ß, protein phosphatase 1, or protein phosphatase 2A, but reduces p35 subunit of Cdk5. Egr-1 silencing also reduces levels of ß-secretase 1 (BACE-1) and BACE-1-cleaved amyloid precursor protein (APP) metabolites (secreted APPß, C99, Aß40, and Aß42) but has no effect on presenilin 1 and presenilin 2. In hippocampal primary neurons, Egr-1 binds to BACE-1 and p35 promoters, enhances tau phosphorylation, activates Cdk5 and BACE-1, and accelerates amyloidogenic APP processing. Blocking Cdk5 action blocks Egr-1-induced tau phosphorylation but has no effect on BACE-1 activation and amyloidogenic APP processing. Blocking BACE-1 action, on the other hand, blocks Egr-1-induced amyloidogenic APP processing but does not affect tau phosphorylation. Egr-1 regulates tau phosphorylation and Aß synthesis in the brain by respectively controlling activities of Cdk5 and BACE-1, suggesting that Egr-1 is a potential therapeutic candidate for the treatment of AD.


Subject(s)
Alzheimer Disease/metabolism , Cognition/physiology , Early Growth Response Protein 1/metabolism , Hippocampus/metabolism , Maze Learning/physiology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Cyclin-Dependent Kinase 5/metabolism , Disease Models, Animal , Early Growth Response Protein 1/genetics , Gene Silencing , Hippocampus/pathology , Mice , Mice, Transgenic , Neurofibrillary Tangles/genetics , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neurons/metabolism , Neurons/pathology , Phosphorylation , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/psychology , Presenilins/metabolism , RNA, Small Interfering , tau Proteins/metabolism
6.
J Neurochem ; 142(1): 56-73, 2017 07.
Article in English | MEDLINE | ID: mdl-28369888

ABSTRACT

Post-synaptic dendritic spines are structurally composed of actin cytoskeleton, which undergoes dynamic morphological changes to accommodate incoming synaptic activity. Drebrin is an actin-binding protein highly expressed in dendritic spines that serves an important role in regulating spine morphology. Functionally, loss of drebrin directly correlates with deficits in learning and memory, as is the case observed in Alzheimer's disease. Despite these findings, the regulatory factor responsible for drebrin loss remains unclear. Here, we show that early growth response-1 (Egr-1), an inducible zinc finger transcription factor, down-regulates drebrin expression. Chromatin immunoprecipitation analyses identified Egr-1 binding sites upstream of the drebrin start site in neuronal cells. Over-expression of Egr-1 in vitro in primary hippocampal neurons or in vivo in homogenates prepared from the hippocampi of an inducible mouse model of Egr-1 show reduced drebrin mRNA and protein levels. Conversely, increased drebrin was detected in hippocampal samples isolated from Egr-1-deficient brain. These data demonstrate that Egr-1 interacts with the drebrin promoter and negatively regulates drebrin expression. Furthermore, immunocytochemical and Golgi staining analyses revealed reduced drebrin protein and dendritic spine density as well as reduced expression of synaptic markers in in vitro hippocampal neurons over-expressing Egr-1 and in vivo inducible mouse model of Egr-1. In contrast, increased drebrin expression correlated with increased dendritic spine density was detected in samples from Egr-1-deficient mice. These data provide evidence that Egr-1 is a novel regulator of drebrin expression, which is linked to changes in dendritic spine density.


Subject(s)
Dendritic Spines/physiology , Early Growth Response Protein 1/metabolism , Neuropeptides/biosynthesis , Animals , Binding Sites , Cell Line, Tumor , Down-Regulation , Early Growth Response Protein 1/genetics , Female , Hippocampus/cytology , Hippocampus/growth & development , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/genetics , Primary Cell Culture , Promoter Regions, Genetic/genetics
7.
Neurobiol Dis ; 103: 78-88, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28396259

ABSTRACT

Progressive accumulation of amyloid-ß peptide (Aß) in the brain is implicated as the central event in the development of Alzheimer's disease (AD). It is thought that extracellular Aß triggers toxic signals leading to neurodegeneration. The events downstream of Aß however are not entirely clear. Clusterin (Apo J) is one of the major risk factors for sporadic form of AD. Clusterin binds to Aß and prevents Aß aggregation. In addition, clusterin promotes Aß degradation and accelerates Aß clearance from the brain. Clusterin thus protects neurons from Aß and loss of clusterin level in the brain is implicated as promoting AD pathology. In this study, we found that the level of clusterin protein but not mRNA is reduced in the brains of 3xTg-AD mice. When rat hippocampal primary neurons were treated with Aß1-42, level of clusterin protein but not mRNA was downregulated. Aß1-42-induced downregulation of clusterin was blocked by lysosome inhibitors bafilomycin A1 and ammonium chloride. In neurons, Aß1-42 induced expression of sortilin, a lysosomal sorting protein that targets proteins to lysosome for degradation. In BE(2) M17 human neuroblastoma cells, clusterin bound to sortilin and when sortilin expression was silenced, Aß1-42-induced clusterin downregulation was almost completely blocked. Our data demonstrate that in neurons, Aß1-42 promotes lysosomal degradation of clusterin by inducing expression of sortilin and provide a novel mechanism by which Aß promotes AD pathogenesis.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Amyloid beta-Peptides/toxicity , Clusterin/metabolism , Hippocampus/metabolism , Lysosomes/metabolism , Neurons/metabolism , Peptide Fragments/toxicity , Animals , Cells, Cultured , Female , Hippocampus/drug effects , Hippocampus/pathology , Humans , Lysosomes/drug effects , Lysosomes/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/pathology , Rats
8.
J Biol Chem ; 291(42): 22276-22287, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27576688

ABSTRACT

Accumulation of amyloid-ß peptide (Aß) in the brain is regarded as central to Alzheimer's disease (AD) pathogenesis. Aß is generated by a sequential cleavage of amyloid precursor protein (APP) by ß-secretase 1 (BACE-1) followed by γ-secretase. BACE-1 cleavage of APP is the committed step in Aß synthesis. Understanding the mechanism by which BACE-1 is activated leading to Aß synthesis in the brain can provide better understanding of AD pathology and help to develop novel therapies. In this study, we found that the levels of Aß and BACE-1 are significantly reduced in the brains of mice lacking transcription factor early growth response 1 (Egr-1) when compared with the WT. We demonstrate that in COS-7 cells, Egr-1 binds to the BACE-1 promoter and activates BACE-1 transcription. In rat hippocampal primary neurons, overexpression of Egr-1 induces BACE-1 expression, activates BACE-1, promotes amyloidogenic APP processing, and enhances Aß synthesis. In mouse hippocampal primary neurons, knockdown of BACE-1 almost completely blocks Egr-1-induced amyloidogenic APP processing and Aß synthesis. Our data indicate that Egr-1 promotes Aß synthesis via transcriptional activation of BACE-1 and suggest that Egr-1 plays role in activation of BACE-1 and acceleration of Aß synthesis in AD brain. Egr-1 is a potential therapeutic target for AD.


Subject(s)
Amyloid Precursor Protein Secretases/biosynthesis , Aspartic Acid Endopeptidases/biosynthesis , Early Growth Response Protein 1/metabolism , Gene Expression Regulation, Enzymologic , Hippocampus/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , COS Cells , Chlorocebus aethiops , Early Growth Response Protein 1/genetics , Hippocampus/pathology , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Rats , Trans-Activators/genetics
9.
PLoS One ; 11(8): e0160635, 2016.
Article in English | MEDLINE | ID: mdl-27548710

ABSTRACT

Microtubule-associated protein tau is the major component of paired helical filaments (PHFs) associated with the neuropathology of Alzheimer's disease (AD). Tau in the normal brain binds and stabilizes microtubules. Tau isolated from PHFs is hyperphosphorylated, which prevents it from binding to microtubules. Tau phosphorylation has been suggested to be involved in the development of NFT pathology in the AD brain. Recently, we showed that 14-3-3ζ is bound to tau in the PHFs and when incubated in vitro with 14-3-3ζ, tau formed amorphous aggregates, single-stranded straight filaments, double stranded ribbon-like filaments and PHF-like filaments that displayed close resemblance with corresponding ultrastructures of AD brain. Surprisingly however, phosphorylated and non-phosphorylated tau aggregated in a similar manner, indicating that tau phosphorylation does not affect in vitro tau aggregation (Qureshi et al (2013) Biochemistry 52, 6445-6455). In this study, we have examined the role of tau phosphorylation in tau aggregation in cellular level. We have found that in human M17 neuroblastoma cells, tau phosphorylation by GSK3ß or PKA does not cause tau aggregation, but promotes 14-3-3ζ-induced tau aggregation by destabilizing microtubules. Microtubule disrupting drugs also promoted 14-3-3ζ-induced tau aggregation without changing tau phosphorylation in M17 cell. In vitro, when incubated with 14-3-3ζ and microtubules, nonphosphorylated tau bound to microtubules and did not aggregate. Phosphorylated tau on the other hand did not bind to microtubules and aggregated. Our data indicate that microtubule-bound tau is resistant to 14-3-3ζ-induced tau aggregation and suggest that tau phosphorylation promotes tau aggregation in the brain by detaching tau from microtubules and thus making it accessible to 14-3-3ζ.


Subject(s)
14-3-3 Proteins/metabolism , Microtubules/metabolism , Neurons/metabolism , Protein Aggregates , tau Proteins/metabolism , 14-3-3 Proteins/genetics , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Microtubules/ultrastructure , Neurons/ultrastructure , Phosphorylation , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , tau Proteins/genetics
10.
J Biol Chem ; 290(49): 29603-16, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26475861

ABSTRACT

The N-methyl-d-aspartate receptor (NMDAR) controls synaptic plasticity and memory function and is one of the major inducers of transcription factor Egr-1 in the hippocampus. However, how Egr-1 mediates the NMDAR signal in neurons has remained unclear. Here, we show that the hippocampus of mice lacking Egr-1 displays electrophysiology properties and ultrastructure that are similar to mice overexpressing PSD-95, a major scaffolding protein of postsynaptic density involved in synapse formation, synaptic plasticity, and synaptic targeting of AMPA receptors (AMPARs), which mediate the vast majority of excitatory transmission in the CNS. We demonstrate that Egr-1 is a transcription repressor of the PSD-95 gene and is recruited to the PSD-95 promoter in response to NMDAR activation. Knockdown of Egr-1 in rat hippocampal primary neurons blocks NMDAR-induced PSD-95 down-regulation and AMPAR endocytosis. Likewise, overexpression of Egr-1 in rat hippocampal primary neurons causes reduction in PSD-95 protein level and promotes AMPAR endocytosis. Our data indicate that Egr-1 is involved in NMDAR-mediated PSD-95 down-regulation and AMPAR endocytosis, a process important in the expression of long term depression.


Subject(s)
Early Growth Response Protein 1/metabolism , Guanylate Kinases/metabolism , Hippocampus/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , COS Cells , Chlorocebus aethiops , Disks Large Homolog 4 Protein , Electrophysiology , Endocytosis , Humans , Long-Term Synaptic Depression , Memory , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Neuronal Plasticity , Promoter Regions, Genetic , Rats , Signal Transduction
11.
Arch Biochem Biophys ; 558: 28-35, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24956593

ABSTRACT

Protein phosphatase 1 (PP1) is one of the major Ser/Thr phosphatases in mammalian cells. There are four isoforms of PP1 namely, PP1α, PP1ß/δ, PP1γ1 and PP1γ2. PP1γ and PP1ß translocate to the nucleus by binding to a co-transporter that contains a nuclear localization signal. The mechanism by which PP1α shuttles between the nucleus and the cytosol is not known. In this study, we found that PP1α co-immunoprecipitates with 14-3-3ζ from HEK-293 cell lysates. By co-immunoprecipitation and GST pull-down assay, we determined that 14-3-3ζ binds to both PP1α (WT) and PP1α (T320A), and that phosphorylation of PP1α is not required for binding. Using PP1α deletion mutants, we located the 14-3-3ζ binding region within PP1α residues 159-279. An in vitro assay showed that 14-3-3ζ does not affect PP1α activity. When HEK-293 cells expressing PP1α and 14-3-3ζ were subjected to subcellular fractionation, the ratio of cytosolic vs. nuclear PP1α was significantly higher in cells expressing PP1α and 14-3-3ζ than those expressing PP1α alone. In cells expressing a dominant negative 14-3-3ζ (K49E), PP1α accumulated in the nucleus. Our results show that 14-3-3ζ binds to PP1α and causes its retention in the cytosol which suggests that 14-3-3ζ regulates nuclear trafficking of PP1α in mammalian cells.


Subject(s)
14-3-3 Proteins/metabolism , Cell Nucleus/metabolism , Protein Phosphatase 1/metabolism , 14-3-3 Proteins/genetics , Active Transport, Cell Nucleus , Binding Sites , Cytosol/metabolism , HEK293 Cells , Humans , Phosphorylation
12.
PLoS One ; 8(12): e84615, 2013.
Article in English | MEDLINE | ID: mdl-24367683

ABSTRACT

b-Amyloid peptide accumulation, tau hyperphosphorylation, and synapse loss are characteristic neuropathological symptoms of Alzheimer's disease (AD). Tau hyperphosphorylation is suggested to inhibit the association of tau with microtubules, making microtubules unstable and causing neurodegeneration. The mechanism of tau phosphorylation in AD brain, therefore, is of considerable significance. Although PHF-tau is phosphorylated at over 40 Ser/Thr sites, Ser(262) phosphorylation was shown to mediate b-amyloid neurotoxicity and formation of toxic tau lesions in the brain. In vitro, PKA is one of the kinases that phosphorylates tau at Ser(262), but the mechanism by which it phosphorylates tau in AD brain is not very clear. 14-3-3z is associated with neurofibrillary tangles and is upregulated in AD brain. In this study, we show that 14-3-3z promotes tau phosphorylation at Ser(262) by PKA in differentiating neurons. When overexpressed in rat hippocampal primary neurons, 14-3-3z causes an increase in Ser(262) phosphorylation, a decrease in the amount of microtubule-bound tau, a reduction in the amount of polymerized microtubules, as well as microtubule instability. More importantly, the level of pre-synaptic protein synaptophysin was significantly reduced. Downregulation of synaptophysin in 14-3-3z overexpressing neurons was mitigated by inhibiting the proteosome, indicating that 14-3-3z promotes proteosomal degradation of synaptophysin. When 14-3-3z overexpressing neurons were treated with the microtubule stabilizing drug taxol, tau Ser(262) phosphorylation decreased and synaptophysin level was restored. Our data demonstrate that overexpression of 14-3-3z accelerates proteosomal turnover of synaptophysin by promoting the destabilization of microtubules. Synaptophysin is involved in synapse formation and neurotransmitter release. Our results suggest that 14-3-3z may cause synaptic pathology by reducing synaptophysin levels in the brains of patients suffering from AD.


Subject(s)
14-3-3 Proteins/metabolism , Alzheimer Disease/metabolism , Gene Expression Regulation/physiology , Hippocampus/cytology , Molecular Chaperones/metabolism , Neurons/metabolism , Synaptophysin/metabolism , tau Proteins/metabolism , Analysis of Variance , Animals , Cloning, Molecular , DNA Primers/genetics , Gene Expression Regulation/drug effects , Image Processing, Computer-Assisted , Immunohistochemistry , Microscopy, Fluorescence , Microtubules/drug effects , Paclitaxel/pharmacology , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Rats
13.
Biochemistry ; 52(37): 6445-55, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23962087

ABSTRACT

Alzheimer's disease (AD) is characterized by the presence of abnormal, straight filaments and paired helical filaments (PHFs) that are coated with amorphous aggregates. When PHFs are treated with alkali, they untwist and form filaments with a ribbonlike morphology. Tau protein is the major component of all of these ultrastructures. 14-3-3ζ is present in NFTs and is significantly upregulated in AD brain. The molecular basis of the association of 14-3-3ζ within NFTs and the pathological significance of its association are not known. In this study, we have found that 14-3-3ζ is copurified and co-immunoprecipitates with tau from NFTs of AD brain extract. In vitro, tau binds to both phosphorylated and nonphosphorylated tau. When incubated with 14-3-3ζ, tau forms amorphous aggregates, single-stranded, straight filaments, ribbonlike filaments, and PHF-like filaments, all of which resemble the corresponding ultrastructures found in AD brain. Immuno-electron microscopy determined that both tau and 14-3-3ζ are present in these ultrastructures and that they are formed in an incubation time-dependent manner. Amorphous aggregates are formed first. As the incubation time increases, the size of amorphous aggregates increases and they are incorporated into single-stranded filaments. Single-stranded filaments laterally associate to form double-stranded, ribbonlike, and PHF-like filaments. Both tau and phosphorylated tau aggregate in a similar manner when they are incubated with 14-3-3ζ. Our data suggest that 14-3-3ζ has a role in the fibrillization of tau in AD brain, and that tau phosphorylation does not affect 14-3-3ζ-induced tau aggregation.


Subject(s)
14-3-3 Proteins/metabolism , Alzheimer Disease/pathology , Neurofibrillary Tangles/pathology , tau Proteins/metabolism , Humans , Neurofibrillary Tangles/ultrastructure , Phosphorylation
15.
J Biol Chem ; 286(23): 20569-81, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21489990

ABSTRACT

In the normal brain, tau protein is phosphorylated at a number of proline- and non-proline directed sites, which reduce tau microtubule binding and thus regulate microtubule dynamics. In Alzheimer disease (AD), tau is abnormally hyperphosphorylated, leading to neurofibrillary tangle formation and microtubule disruption, suggesting a loss of regulatory mechanisms controlling tau phosphorylation. Early growth response 1 (Egr-1) is a transcription factor that is significantly up-regulated in AD brain. The pathological significance of this up-regulation is not known. In this study, we found that lentivirus-mediated overexpression of Egr-1 in rat brain hippocampus and primary neurons in culture activates proline-directed kinase Cdk5, inactivates PP1, promotes tau phosphorylation at both proline-directed Ser(396/404) and non-proline-directed Ser(262) sites, and destabilizes microtubules. Furthermore, in Egr-1(-/-) mouse brain, Cdk5 activity was decreased, PP1 activity was increased, and tau phosphorylation was reduced at both proline-directed and non-proline-directed sites. By using nerve growth factor-exposed PC12 cells, we determined that Egr-1 activates Cdk5 to promote phosphorylation of tau and inactivates PP1 via phosphorylation. When Cdk5 was inhibited, tau phosphorylation at both proline- and non-proline directed sites and PP1 phosphorylation were blocked, indicating that Egr-1 acts through Cdk5. By using an in vitro kinase assay and HEK-293 cells transfected with tau, PP1, and Cdk5, we found that Cdk5 phosphorylates Ser(396/404) directly. In addition, by phosphorylating and inactivating PP1, Cdk5 promotes tau phosphorylation at Ser(262) indirectly. Our results indicate that Egr-1 is an in vivo regulator of tau phosphorylation and suggest that in AD brain increased levels of Egr-1 aberrantly activate an Egr-1/Cdk5/PP1 pathway, leading to accumulation of hyperphosphorylated tau, thus destabilizing the microtubule cytoskeleton.


Subject(s)
Early Growth Response Protein 1/metabolism , Hippocampus/metabolism , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Early Growth Response Protein 1/genetics , Enzyme Activation/genetics , Female , HEK293 Cells , Humans , Mice , Mice, Knockout , Microtubules/genetics , Microtubules/metabolism , PC12 Cells , Phosphorylation/genetics , Rats , Rats, Long-Evans , Receptors, Neuropeptide Y , tau Proteins/genetics
16.
J Biol Chem ; 286(7): 5055-68, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21127069

ABSTRACT

In Parkinson disease (PD) brain, a progressive loss of dopaminergic neurons leads to dopamine depletion in the striatum and reduced motor function. Lewy bodies, the characteristic neuropathological lesions found in the brain of PD patients, are composed mainly of α-synuclein protein. Three point mutations in the α-synuclein gene are associated with familial PD. In addition, genome-wide association studies indicate that α-synuclein and Tau protein synergistically increase disease susceptibility in the human population. To determine the mechanism by which α-synuclein and Tau act together, we have used PD-causing neurotoxin MPTP and pathogenic α-synuclein mutants A30P, E46K, and A53T as models. We found that exposure of human neuroblastoma M17 cells to MPTP enhances the intracellular α-synuclein protein level, stimulates Tau protein phosphorylation at Ser(262), and induces apoptosis. In mouse brain, ablation of α-synuclein function significantly suppresses Tau phosphorylation at Ser(262). In vitro, α-synuclein binds to phosphorylated Ser(214) of Tau and stimulates PKA-catalyzed Tau phosphorylation at Ser(262). PD-associated α-synuclein mutations increase α-synuclein binding to Tau and stimulate Tau phosphorylation at Ser(262). In HEK-293 cells, α-synuclein and its all PD-associated mutants destabilize the microtubule cytoskeleton in a similar extent. In contrast, when co-expressed with Tau, these PD-associated mutants destabilize microtubules with significantly higher potency than WT. Our results demonstrate that α-synuclein is an in vivo regulator of Tau protein phosphorylation at Ser(262) and suggest that PD-associated risk factors such as environmental toxins and α-synuclein mutations promote Tau phosphorylation at Ser(262), causing microtubule instability, which leads to loss of dopaminergic neurons in PD brain.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Microtubules/metabolism , Mutation, Missense , Neurotoxins/pharmacology , alpha-Synuclein/metabolism , tau Proteins/metabolism , Amino Acid Substitution , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Genome-Wide Association Study , HEK293 Cells , Humans , Microtubules/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , alpha-Synuclein/genetics , tau Proteins/genetics
17.
J Neurosci ; 29(27): 8743-51, 2009 Jul 08.
Article in English | MEDLINE | ID: mdl-19587281

ABSTRACT

The cause of the conformational change of normal cellular prion protein (PrP) into its disease-associated form is unknown. Posttranslational modifications, such as glycosylation, acetylation, S-nitrosylation, and phosphorylation, are known to induce protein conformational changes. Here, we investigated whether phosphorylation could induce the conformational change of PrP because PrP contains several kinase motifs and has been found recently in the cytosol, in which kinases generally reside. Neuronal cyclin-dependent kinase 5 (Cdk5) phosphorylated recombinant PrP(23-231) at serine 43 (S43) in an in vitro kinase assay. Cdk5-phosphorylated PrP became proteinase K resistant, formed Congo Red-positive fibrils, and formed aggregates that were immunostained with anti-PrP and anti-phospho-PrP(S43) (anti-pPrP(S43)). pPrP(S43) was detected in PrP/Cdk5/p25 cotransfected N2a cells. Roscovitine inhibition of Cdk5 activity or transfection of N2a cells with mutant PrP S43A eliminated the anti-pPrP(S43)-immunopositive protein. Alkaline phosphatase-sensitive and proteinase K-resistant pPrP(S43) immunoreactivity was observed in scrapie-infected but not control-injected mice brains. These results raise the possibility that phosphorylation could represent a physiological mechanism of PrP conversion in vivo.


Subject(s)
Peptide Fragments/metabolism , Prions/metabolism , Serine/metabolism , Animals , Cattle , Cell Line, Tumor , Humans , Mice , Mice, Inbred C57BL , Peptide Fragments/chemistry , Peptide Fragments/genetics , Phosphorylation/genetics , Prions/chemistry , Prions/genetics , Protein Conformation , Serine/chemistry , Serine/genetics
18.
J Neurochem ; 110(2): 719-33, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19457084

ABSTRACT

Proteosomal degradation of proteins is one of the major mechanisms of intracellular protein turnover. Failure of the proteosome to degrade misfolded protein is implicated in the accumulation of alpha-synuclein in Parkinson's disease (PD). Heme oxygenase-1 (HO-1), an enzyme that converts heme to free iron, carbon monoxide (CO) and biliverdin (bilirubin precursor) is expressed in response to various stressors. HO-1 is up-regulated in PD- and Alzheimer's disease-affected neural tissues. In this study, we found that HO-1 over-expression engenders dose-dependent decreases in alpha-synuclein protein levels in human neuroblastoma M17 cells. When over-expression of HO-1 was silenced in HO-1 transfected cells, level of alpha-synuclein was restored. Likewise, treatment of HO-1 over-expressing cells with the HO-1 inhibitor, tin mesoporphyrin, the iron chelator deferoxamine or antagonist of CO-dependent cGMP activation, methylene blue, mitigated the HO-1-induced reduction in alpha-synuclein levels. Furthermore, when HO-1 over-expressing cells were treated with the proteosome inhibitors, lactacystin and MG132, level of alpha-synuclein was almost completely restored. In contrast to the effect on alpha-synuclein [wild-type (WT)] levels, HO-1 over-expression did not significantly impact PD-associated alpha-synuclein (A30P) levels in these cells. HO-1 also significantly reduced aggregation of alpha-synuclein (WT) but not that of A30P. Our results suggest that HO-1, which is expressed when neurons are exposed to toxic stimuli capable of inducing protein misfolding, triggers proteosomal degradation of proteins and prevents intracellular accumulation of protein aggregates and inclusions. Resistance to HO-1 induced proteosomal degradation may render the familial PD-associated A30P mutation prone to toxic intracellular aggregation.


Subject(s)
Gene Expression Regulation, Enzymologic/genetics , Heme Oxygenase-1/genetics , Neuroblastoma/metabolism , Parkinson Disease/metabolism , Proteasome Endopeptidase Complex/metabolism , alpha-Synuclein/metabolism , Animals , Gene Silencing , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/deficiency , Humans , Mutation , Neuroblastoma/enzymology , Neuroblastoma/genetics , Parkinson Disease/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/physiology , Protein Denaturation/genetics , Protein Stability , Rats , Tumor Cells, Cultured , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/genetics
19.
J Biol Chem ; 284(20): 13422-13433, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19304664

ABSTRACT

In Alzheimer disease (AD), frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) and other tauopathies, tau accumulates and forms paired helical filaments (PHFs) in the brain. Tau isolated from PHFs is phosphorylated at a number of sites, migrates as approximately 60-, 64-, and 68-kDa bands on SDS-gel, and does not promote microtubule assembly. Upon dephosphorylation, the PHF-tau migrates as approximately 50-60-kDa bands on SDS-gels in a manner similar to tau that is isolated from normal brain and promotes microtubule assembly. The site(s) that inhibits microtubule assembly-promoting activity when phosphorylated in the diseased brain is not known. In this study, when tau was phosphorylated by Cdk5 in vitro, its mobility shifted from approximately 60-kDa bands to approximately 64- and 68-kDa bands in a time-dependent manner. This mobility shift correlated with phosphorylation at Ser(202), and Ser(202) phosphorylation inhibited tau microtubule-assembly promoting activity. When several tau point mutants were analyzed, G272V, P301L, V337M, and R406W mutations associated with FTDP-17, but not nonspecific mutations S214A and S262A, promoted Ser(202) phosphorylation and mobility shift to a approximately 68-kDa band. Furthermore, Ser(202) phosphorylation inhibited the microtubule assembly-promoting activity of FTDP-17 mutants more than of WT. Our data indicate that FTDP-17 missense mutations, by promoting phosphorylation at Ser(202), inhibit the microtubule assembly-promoting activity of tau in vitro, suggesting that Ser(202) phosphorylation plays a major role in the development of NFT pathology in AD and related tauopathies.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Mutation, Missense , tau Proteins/metabolism , Animals , Cattle , Chromosomes, Human, Pair 17/chemistry , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 17/metabolism , Cyclin-Dependent Kinase 5/chemistry , Cyclin-Dependent Kinase 5/genetics , Humans , Microtubules/genetics , Phosphorylation , Tauopathies/genetics , Tauopathies/metabolism , tau Proteins/chemistry , tau Proteins/genetics
20.
Neurochem Int ; 54(1): 14-27, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18992292

ABSTRACT

FTDP-17 missense tau mutations: G272V, P301L, V337M and R406W promote tau phosphorylation in human and transgenic mice brains by interfering with the tau phosphorylation/dephosphorylation balance. The effect of FTDP-17 mutations on tau phosphorylation by different kinases has been studied previously. However, it is not known how various FTDP-17 mutations affect tau dephosphorylation by phosphoprotein phosphatases. In this study we have observed that when transfected into HEK-293 cells, tau is phosphorylated on various sites that are also phosphorylated in diseased human brains. When transfected cells are lysed and incubated, endogenously phosphorylated tau is dephosphorylated by cellular protein phosphatase 1 (PP1), phosphatase 2A (PP2A) and phosphatase 2B (PP2B), which are also present in the lysate. By using this assay and specific inhibitors of PP1, PP2A and PP2B, we have observed that the G272V mutation promotes tau dephosphorylation by PP2A at Ser(396/404), Ser(235), Thr(231), Ser(202/205) and Ser(214) and by PP2B at Ser(214) but inhibits dephosphorylation by PP2B at Ser(396/404). The P301L mutation promotes tau dephosphorylation at Thr(231) by PP1 and at Ser(396/404), Thr(231), Ser(235) and Ser(202/205) by PP2A but inhibits dephosphorylation at Ser(214) by PP2B. The V337M mutation promotes tau dephosphorylation at Ser(235), Thr(231) and Ser(202/205) by PP2A and at Ser(202/205) by PP2B whereas the R406W mutation promotes tau dephosphorylation at Ser(396/404) by PP1, PP2A and PP2B but inhibits dephosphorylation at Ser(202/205) and Ser(235) by PP1 and PP2A, respectively. Our results indicate that each FTDP-17 tau mutation not only site-specifically inhibits tau dephosphorylation on some sites but also promotes dephosphorylation by phosphatases on other sites.


Subject(s)
Mutation, Missense , Phosphoprotein Phosphatases/metabolism , tau Proteins/metabolism , Amino Acids/metabolism , Antibodies, Monoclonal , Brain/metabolism , Cell Line , Cloning, Molecular , DNA, Complementary/genetics , Humans , Kidney , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation , Recombinant Proteins/metabolism , Transfection , tau Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...