Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Alzheimers Dis ; 73(1): 147-161, 2020.
Article in English | MEDLINE | ID: mdl-31771053

ABSTRACT

Tau oligomers have been shown to transmit tau pathology from diseased neurons to healthy neurons through seeding, tau misfolding, and aggregation that is thought to play an influential role in the progression of Alzheimer's disease (AD) and related tauopathies. To develop a small molecule therapeutic for AD and related tauopathies, we have developed in vitro and cellular assays to select molecules inhibiting the first step in tau aggregation, the self-association of tau into oligomers. In vivo validation studies of an optimized lead compound were independently performed in the htau mouse model of tauopathy that expresses the human isoforms of tau without inherited tauopathy mutations that are irrelevant to AD. Treated mice did not show any adverse events related to the compound. The lead compound significantly reduced the level of self-associated tau and total and phosphorylated insoluble tau aggregates. The dose response was linear with respect to levels of compound in the brain. A confirmatory study was performed with male htau mice that gave consistent results. The results validated our screening approach by showing that targeting tau self-association can inhibit the entire tau aggregation pathway by using the selected and optimized lead compound whose activity translated from in vitro and cellular assays to an in vivo model of tau aggregation.


Subject(s)
tau Proteins/antagonists & inhibitors , tau Proteins/genetics , Alzheimer Disease/drug therapy , Animals , Brain Chemistry/drug effects , Dose-Response Relationship, Drug , Female , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Immunohistochemistry , Male , Mice , Mice, Transgenic , Mutation/genetics , Sex Characteristics , Small Molecule Libraries , Tauopathies/drug therapy , Tauopathies/genetics , tau Proteins/metabolism
2.
Article in English | MEDLINE | ID: mdl-27379014

ABSTRACT

Type 2 diabetes mellitus is characterized by the deposition of islet amyloid polypeptide (IAPP) as amyloid in islets, a process thought to be toxic to ß-cells. To determine the feasibility of targeting these aggregates therapeutically, we vaccinated transgenic (Tg) mice that overexpress human IAPP and were fed a high-fat diet to promote their diabetic phenotype. Our findings indicate that prophylactic vaccination with IAPP and its derivative IAPP7-19-TT, protects wild-type female mice, but not males, from obesity-induced early mortality, and the derivative showed a strong trend for prolonging the lifespan of Tg females but not males. Furthermore, IAPP7-19-TT-immunized Tg females cleared a glucose bolus more efficiently than controls, while IAPP-immunized Tg females showed an impaired ability to clear a glucose bolus compared to their adjuvant injected Tg controls. Interestingly, IAPP or IAPP7-19-TT treatments had no effect on glucose clearance in Tg males. Overall, these beneficial effects of IAPP targeted immunization depend on Tg status, sex, and immunogen. Hence, future studies in this field should carefully consider these variables that clearly affect the therapeutic outcome. In conclusion, IAPP targeting immunotherapy may have benefits in patients with type 2 diabetes.

3.
J Neurosci ; 34(50): 16835-50, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25505335

ABSTRACT

Antibodies or their derivatives as imaging probes for pathological tau protein have great potential, but have not been well studied. In particular, smaller, single-chain-variable antibody fragments (scFv's) are attractive for detecting tau lesions in live subjects. Here, we generated libraries of scFv's and identified numerous phospho-tau-selective scFv's. Peripheral injection of one of these scFv's consistently resulted in a strong in vivo brain signal in transgenic tauopathy mice, but not in wild-type or amyloid-ß plaque mice. The parent tau antibody provided similar results, albeit with a weaker signal intensity. The imaging signal correlated very well with colocalization of the probe with intraneuronal tau aggregates. Both were associated with markers of endosomes, autophagosomes, and lysosomes, suggesting their interaction in these degradation pathways. Such specific antibody-derived imaging probes have great potential as diagnostic markers for Alzheimer's disease and related tauopathies.


Subject(s)
Diagnostic Imaging , Single-Chain Antibodies/metabolism , tau Proteins/metabolism , Amino Acid Sequence , Animals , Diagnostic Imaging/methods , Female , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Single-Chain Antibodies/genetics , tau Proteins/genetics
4.
Acta Neuropathol Commun ; 1: 34, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-24252661

ABSTRACT

BACKGROUND: Tau is a microtubule stabilizing protein and is mainly expressed in neurons. Tau aggregation into oligomers and tangles is considered an important pathological event in tauopathies, such as frontotemporal dementia (FTD) and Alzheimer's disease (AD). Tauopathies are also associated with deficits in synaptic plasticity such as long-term potentiation (LTP), but the specific role of tau in the manifestation of these deficiencies is not well-understood. We examined long lasting forms of synaptic plasticity in JNPL3 (BL6) mice expressing mutant tau that is identified in some inherited FTDs. RESULTS: We found that aged (>12 months) JNPL3 (BL6) mice exhibit enhanced hippocampal late-phase (L-LTP), while young JNPL3 (BL6) mice (age 6 months) displayed normal L-LTP. This enhanced L-LTP in aged JNPL3 (BL6) mice was rescued with the GABAAR agonist, zolpidem, suggesting a loss of GABAergic function. Indeed, we found that mutant mice displayed a reduction in hippocampal GABAergic interneurons. Finally, we also found that expression of mutant tau led to severe sensorimotor-gating and hippocampus-dependent memory deficits in the aged JNPL3 (BL6) mice. CONCLUSIONS: We show for the first time that hippocampal GABAergic function is impaired by pathological tau protein, leading to altered synaptic plasticity and severe memory deficits. Increased understanding of the molecular mechanisms underlying the synaptic failure in AD and FTD is critical to identifying targets for therapies to restore cognitive deficiencies associated with tauopathies.


Subject(s)
GABAergic Neurons/physiology , Interneurons/physiology , Long-Term Potentiation , Sensory Gating/physiology , Tauopathies/physiopathology , Animals , Cell Count , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Disease Models, Animal , GABA-A Receptor Agonists/pharmacology , GABAergic Neurons/drug effects , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Interneurons/drug effects , Interneurons/pathology , Long-Term Potentiation/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Pyridines/pharmacology , Sensory Gating/drug effects , Tauopathies/drug therapy , Tauopathies/pathology , Zolpidem , tau Proteins/genetics , tau Proteins/metabolism
5.
Neuroimage ; 64: 693-702, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22960250

ABSTRACT

The impairment of axonal transport by overexpression or hyperphosphorylation of tau is well documented for in vitro conditions; however, only a few studies on this phenomenon have been conducted in vivo, using invasive procedures, and with contradictory results. Here we used the non-invasive, Manganese-Enhanced Magnetic Resonance Imaging technique (MEMRI), to study for the first time a pure model of tauopathy, the JNPL3 transgenic mouse line, which overexpresses a mutated (P301L) form of the human tau protein. We show progressive impairment in neuronal transport as tauopathy advances. These findings are further supported by a significant correlation between the severity of the impairment in neuronal transport assessed by MEMRI, and the degree of abnormal tau assessed by histology. Unlike conventional techniques that focus on axonal transport measurement, MEMRI can provide a global analysis of neuronal transport, i.e. from dendrites to axons and at the macroscopic scale of fiber tracts. Neuronal transport impairment has been shown to be a key pathogenic process in Alzheimer's disease and numerous other neurodegenerative disorders. Hence, MEMRI provides a promising set of functional biomarkers to be used during preclinical trials to facilitate the selection of new drugs aimed at restoring neuronal transport in neurodegenerative diseases.


Subject(s)
Chlorides/pharmacokinetics , Magnetic Resonance Imaging/methods , Manganese Compounds/pharmacokinetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Tauopathies/metabolism , Tauopathies/pathology , Animals , Contrast Media/pharmacokinetics , Humans , Mice , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Synaptic Transmission , tau Proteins/genetics
6.
Front Psychiatry ; 2: 59, 2011.
Article in English | MEDLINE | ID: mdl-22025915

ABSTRACT

Recent studies have shown that immunotherapy clears amyloid beta (Aß) plaques and reduces Aß levels in mouse models of Alzheimer's disease (AD), as well as in AD patients. Tangle pathology is also relevant for the neurodegeneration in AD, and our studies have shown that active immunization with an AD related phospho-tau peptide reduces aggregated tau within the brain and slows the progression of tauopathy-induced behavioral impairments. Thus, clearance of neurofibrillary tangles and/or their precursors may reduce synaptic and neuronal loss associated with AD and other tauopathies. So far the mechanisms involved in antibody-mediated clearance of tau pathology are yet to be elucidated. In this study we have used a mouse brain slice model to examine the uptake and localization of FITC labeled anti-tau antibodies. Confocal microscopy analysis showed that the FITC labeled anti-tau antibody co-stained with phosphorylated tau, had a perinuclear appearance and co-localized with markers of the endosomal/lysosomal pathway. Additionally, tau and FITC-IgG were found together in an enriched lysosome fraction. In summary, antibody-mediated clearance of intracellular tau aggregates appears to occur via the lysosomal pathway.

7.
N Biotechnol ; 28(5): 511-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21473943

ABSTRACT

Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease, Huntington's disease (HD) or amyotrophic lateral sclerosis (ALS) are all characterised histologically by the presence of deposits of misfolded proteins, tau and amyloid-ß, α-synuclein, huntingtin or superoxide dismutase, respectively. Currently, these illnesses do not have any disease modifying treatment options. A novel therapeutic strategy that is being pursued is immunomodulation, which is using the body's immune system to target the self-proteins that are deposited. Most of these promising approaches are still in preclinical development while some have progressed to Phase III clinical trials. As new insights are gained, it is hoped that these immunotherapies will be effective tools at slowing the progression of these debilitating diseases.


Subject(s)
Antibodies/therapeutic use , Neurodegenerative Diseases/drug therapy , Animals , Antibodies/immunology , Disease Progression , Humans , Immunologic Techniques , Immunotherapy , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/pathology
8.
J Neurosci ; 28(48): 12798-807, 2008 Nov 26.
Article in English | MEDLINE | ID: mdl-19036972

ABSTRACT

In Alzheimer's disease, tau is hyperphosphorylated, which is thought to detach it from microtubules (MTs), induce MT destabilization, and promote aggregation. Using a previously described in vivo model, we investigated whether hyperphosphorylation impacts tau function in wild-type and transgenic mice. We found that after anesthesia-induced hypothermia, MT-free tau was hyperphosphorylated, which impaired its ability to bind MTs and promote MT assembly. MT-bound tau was more resistant to hyperphosphorylation compared with free tau and tau did not dissociate from MTs in wild-type mice. However, 3-repeat tau detached from MT in the transgenic mice. Surprisingly, dissociation of tau from MTs did not lead to overt depolymerization of tubulin, and there was no collapse, or disturbance of axonal MT networks. These results indicate that, in vivo, a subpopulation of tau bound to MTs does not easily dissociate under conditions that extensively phosphorylate tau. Tau remaining on the MTs under these conditions is sufficient to maintain MT network integrity.


Subject(s)
Anesthetics/pharmacology , Axons/metabolism , Microtubules/metabolism , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Axons/drug effects , Axons/ultrastructure , Brain/metabolism , Brain/physiopathology , Brain/ultrastructure , Disease Models, Animal , Female , Hypothermia, Induced , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Microtubules/drug effects , Microtubules/ultrastructure , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Binding/genetics , Trinucleotide Repeats/genetics , tau Proteins/drug effects , tau Proteins/genetics
9.
Neurobiol Dis ; 31(1): 46-57, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18504134

ABSTRACT

In the last decade, multiple lines of transgenic APP overexpressing mice have been created that recapitulate certain aspects of Alzheimer's disease (AD). However, none of the previously reported transgenic APP overexpressing rat models developed AD-like beta-amyloid (Abeta) deposits, or age-related learning and memory deficits. In the present study, we have characterized a transgenic rat model overexpressing transgenes with three, familial AD mutations (two in APP and one in PS1) that were developed by Flood et al. [Flood, D.G., et al., Abeta deposition in a transgenic rat model of Alzheimer's disease. Society for Neuroscience 2003, Washington, DC, 2003]. From the age of 9 months, these rats develop Abeta deposits in both diffuse and compact forms, with the latter being closely associated with activated microglia and reactive astrocytes. Impaired long-term potentiation (LTP) was revealed by electrophysiological recordings performed on hippocampal slices from rats at 7 months of age, which is 2 months before the appearance of amyloid plaques. The deficit in LTP was accompanied by impaired spatial learning and memory in the Morris water maze, which became more pronounced in transgenic rats of 13 months of age. For Tg rats of both ages, there was a trend for cognitive impairment to correlate with total Abeta42 levels in the hippocampus. The rat model therefore recapitulates AD-like amyloid pathology and cognitive impairment. The advantage of the rat model over the available mouse models is that rats provide better opportunities for advanced studies, such as serial CSF sampling, electrophysiology, neuroimaging, cell-based transplant manipulations, and complex behavioral testing.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Cognition Disorders/etiology , Neuronal Plasticity/physiology , Plaque, Amyloid/pathology , Alzheimer Disease/complications , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Animals, Genetically Modified , Brain/metabolism , Cognition Disorders/physiopathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Excitatory Postsynaptic Potentials , Immunoblotting , Immunohistochemistry , Male , Maze Learning , Mutation , Organ Culture Techniques , Plaque, Amyloid/metabolism , Presenilins/genetics , Presenilins/metabolism , Rats , Rats, Sprague-Dawley , Synaptic Transmission/physiology
10.
Neuron ; 57(5): 680-90, 2008 Mar 13.
Article in English | MEDLINE | ID: mdl-18341989

ABSTRACT

Cyclin-dependent kinase 5 (cdk5) has been implicated in Alzheimer's disease (AD) pathogenesis. Here, we demonstrate that overexpression of p25, an activator of cdk5, led to increased levels of BACE1 mRNA and protein in vitro and in vivo. A p25/cdk5 responsive region containing multiple sites for signal transducer and activator of transcription (STAT1/3) was identified in the BACE1 promoter. STAT3 interacts with the BACE1 promoter, and p25-overexpressing mice had elevated levels of pSTAT3 and BACE1, whereas cdk5-deficient mice had reduced levels. Furthermore, mice with a targeted mutation in the STAT3 cdk5 responsive site had lower levels of BACE1. Increased BACE levels in p25 overexpressing mice correlated with enhanced amyloidogenic processing that could be reversed by a cdk5 inhibitor. These data demonstrate a pathway by which p25/cdk5 increases the amyloidogenic processing of APP through STAT3-mediated transcriptional control of BACE1 that could have implications for AD pathogenesis.


Subject(s)
Amyloid Precursor Protein Secretases/biosynthesis , Amyloid beta-Protein Precursor/biosynthesis , Cyclin-Dependent Kinase 5/biosynthesis , Nerve Tissue Proteins/biosynthesis , Transcription, Genetic/physiology , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase 5/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , PC12 Cells , Phosphotransferases , Rats
11.
J Neurosci ; 27(12): 3090-7, 2007 Mar 21.
Article in English | MEDLINE | ID: mdl-17376970

ABSTRACT

Postoperative cognitive dysfunction, confusion, and delirium are common after general anesthesia in the elderly, with symptoms persisting for months or years in some patients. Even middle-aged patients are likely to have postoperative cognitive dysfunction for months after surgery, and Alzheimer's disease (AD) patients appear to be particularly at risk of deterioration after anesthesia. Several investigators have thus examined whether general anesthesia is associated with AD, with some studies suggesting that exposure to anesthetics may increase the risk of AD. However, little is known on the biochemical consequences of anesthesia on pathogenic pathways in vivo. Here, we investigated the effect of anesthesia on tau phosphorylation and amyloid precursor protein (APP) metabolism in mouse brain. We found that, regardless of the anesthetic used, anesthesia induced rapid and massive hyperphosphorylation of tau, rapid and prolonged hypothermia, inhibition of Ser/Thr PP2A (protein phosphatase 2A), but no changes in APP metabolism or Abeta (beta-amyloid peptide) accumulation. Reestablishing normothermia during anesthesia completely rescued tau phosphorylation to normal levels. Our results indicate that changes in tau phosphorylation were not a result of anesthesia per se, but a consequence of anesthesia-induced hypothermia, which led to inhibition of phosphatase activity and subsequent hyperphosphorylation of tau. These findings call for careful monitoring of core temperature during anesthesia in laboratory animals to avoid artifactual elevation of protein phosphorylation. Furthermore, a thorough examination of the effect of anesthesia-induced hypothermia on the risk and progression of AD is warranted.


Subject(s)
Anesthesia/adverse effects , Hypothermia/metabolism , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , tau Proteins/metabolism , Anesthetics/administration & dosage , Anesthetics/adverse effects , Animals , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/metabolism , Hypothermia/enzymology , Male , Mice , Phosphorylation/drug effects , Protein Phosphatase 2
12.
Proc Natl Acad Sci U S A ; 102(19): 6990-5, 2005 May 10.
Article in English | MEDLINE | ID: mdl-15867159

ABSTRACT

Neurofibrillary tangles composed of hyperphosphorylated, aggregated tau are a common pathological feature of tauopathies, including Alzheimer's disease. Abnormal phosphorylation of tau by kinases or phosphatases has been proposed as a pathogenic mechanism in tangle formation. To investigate whether kinase inhibition can reduce tauopathy and the degeneration associated with it in vivo, transgenic mice overexpressing mutant human tau were treated with the glycogen synthase kinase-3 (GSK-3) inhibitor lithium chloride. Treatment resulted in significant inhibition of GSK-3 activity. Lithium administration also resulted in significantly lower levels of phosphorylation at several epitopes of tau known to be hyperphosphorylated in Alzheimer's disease and significantly reduced levels of aggregated, insoluble tau. Administration of a second GSK-3 inhibitor also correlated with reduced insoluble tau levels, supporting the idea that lithium exerts its effect through GSK-3 inhibition. Levels of aggregated tau correlated strongly with degree of axonal degeneration, and lithium-chloride-treated mice showed less degeneration if administration was started during early stages of tangle development. These results support the idea that kinases are involved in tauopathy progression and that kinase inhibitors may be effective therapeutically.


Subject(s)
Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Lithium Chloride/pharmacology , Animals , Disease Progression , Epitopes , Humans , Image Processing, Computer-Assisted , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Lithium/chemistry , Mice , Mice, Transgenic , Neurodegenerative Diseases/pathology , Neurons/pathology , Phosphorylation , Tauopathies , tau Proteins/chemistry
13.
Neuroreport ; 15(14): 2223-6, 2004 Oct 05.
Article in English | MEDLINE | ID: mdl-15371738

ABSTRACT

Tau is a microtubule-associated protein involved in axonal elongation and central to the pathogenesis of a number of neurodegenerative conditions. To better establish the contribution of the cellular context to tau-dependent microtubule organization, we compared the phenotypes resulting from heterologous tau expression in different mammalian cell lines after disruption of the actin cytoskeleton. After cytochalasin D treatment, tau-expressing CHO cells display one or two long neurite-like extensions whereas cells transfected with MAP2c developed multiple shorter processes. By contrast, under the same conditions, tau-transfected PtK2 cells elaborate microtubule bundles forming numerous processes. These results suggest that cell-specific factors are involved in tau-dependent microtubule organization, a notion that could facilitate functional assessment of tau abnormalities associated with neurodegenerative disease.


Subject(s)
Microtubules/physiology , tau Proteins/physiology , Animals , CHO Cells , Cell Line , Cricetinae , Humans , Neurons/cytology , Neurons/physiology
14.
J Neurosci Res ; 76(1): 110-20, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15048935

ABSTRACT

Cyclin-dependent kinase-5 (CDK5), a unique CDK family member, is active primarily in the central nervous system (CNS). Previous studies suggest that CDK5 is proapoptotic and contributes to tau hyperphosphorylation and neurodegeneration in Alzheimer's disease. The objective of this study was to examine CDK5 effects on apoptotic progression and tau phosphorylation. Immortalized embryonic mouse brain cortical cells were used to establish a stable cell line that overexpressed wild-type human tau. In these studies, thapsigargin, which induces endoplasmic reticulum stress and can cause accumulation of misfolded proteins, was used to induce apoptosis. Caspase-3 activity and poly-(ADP-ribose)-polymerase (PARP) cleavage, as measures of apoptosis, were significantly increased 24 and 48 hr after thapsigargin treatment, and these events were unaffected by tau expression. Although transient coexpression of CDK5 and its activator, p25, increased CDK5 activity greater than tenfold, increases in caspase-3 activity in response to thapsigargin treatment were unaffected by the presence of CDK5/p25. Tau phosphorylation at the PHF-1 epitope, but not the Tau-1 epitope, was increased significantly in CDK5/p25-transfected cells compared to cells transfected with dominant negative CDK5 (DNCDK5). The PHF-1 epitope remained phosphorylated until 48 hr after thapsigargin treatment in the CDK5/p25-transfected cells. Over the course of apoptosis in this model, phosphorylation of the Tau-1 epitope was unaffected in cells transfected with DNCDK5, vector, or CDK5/p25. In summary, these results demonstrate that CDK5 does not have a significant impact on tau phosphorylation and thapsigargin-induced apoptosis in this neuronal cell model.


Subject(s)
Apoptosis/physiology , Cerebral Cortex/physiology , Cyclin-Dependent Kinases/physiology , Neurons/physiology , tau Proteins/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Blotting, Western , Caspase 3 , Caspases/metabolism , Cell Line , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Chromobox Protein Homolog 5 , Cyclin-Dependent Kinase 5 , Doxycycline/pharmacology , Enzyme Activation/drug effects , Enzyme Activators/pharmacology , Epitopes/drug effects , Epitopes/physiology , Humans , Mice , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Phosphorylation , Plasmids/genetics , Thapsigargin/pharmacology , Transfection
15.
J Biol Chem ; 279(9): 7893-900, 2004 Feb 27.
Article in English | MEDLINE | ID: mdl-14660557

ABSTRACT

Frontotemporal dementia and Parkinsonism linked to chromosome 17 (FTDP-17) is an autosomal dominant neurodegenerative disorder caused by mutations in the gene that encodes for tau, a microtubule-binding protein. Neuropathologically the disease is characterized by extensive neuronal loss in the frontal and temporal lobes and the filamentous accumulation of hyperphosphorylated tau. The R406W missense mutation was originally described in an American and a Dutch family. Although R406W tau is hyperphosphorylated in FTDP-17 cases, R406W tau expressed in cell model systems has not shown increased phosphorylation. The purpose of this study was to establish a neuronal model system in which the phosphorylation of R406W tau is increased and thus more representative of the in vivo situation. To accomplish this goal immortalized mouse cortical cells that express low levels of endogenous tau were stably transfected with human wild type or R406W tau. In this neuronal model R406W tau was more highly phosphorylated at numerous epitopes and showed decreased microtubule binding compared with wild type tau, an effect that could be reversed by dephosphorylation. In addition the expression of R406W tau in the cortical cells resulted in increased cell death as compared with wild type tau-expressing cells when the cells were exposed to an apoptotic stressor. These results indicate that in an appropriate cellular context R406W tau is hyperphosphorylated, which leads to decreased microtubule binding. Furthermore, expression of R406W tau sensitized cells to apoptotic stress, which may contribute to the neuronal cell loss that occurs in this FTDP-17 tauopathy.


Subject(s)
Cerebral Cortex/ultrastructure , Microtubules/metabolism , Mutation, Missense , Neurons/ultrastructure , tau Proteins/genetics , tau Proteins/metabolism , Animals , Apoptosis/genetics , Caspase 3 , Caspases/metabolism , Cell Line, Transformed , Gene Expression , Humans , Mice , Neurons/physiology , Osmotic Pressure , Phosphorylation , Transfection , tau Proteins/chemistry
16.
J Neurochem ; 81(2): 307-13, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12064478

ABSTRACT

Cyclin dependent kinase 5 (Cdk5) is a proline-direct protein kinase that is most active in the CNS, and has been implicated as a contributing factor in certain neurodegenerative diseases. Further, there is evidence to suggest that Cdk5 may facilitate the progression of apoptosis. However, the mechanisms involved have not been elucidated. The tumor suppressor protein p53, a transcription factor that is regulated by phosphorylation, increases the expression of genes that control growth arrest or cell death. To understand how Cdk5 could facilitate apoptosis, the effects of Cdk5 on p53 activity were examined. In the present study it is shown that in apoptotic PC12 cells the levels of p53 and Cdk5 increase concomitantly. Further, Cdk5/p25 effectively phosphorylates recombinant p53 in vitro. Transient transfection of Cdk5/p25 into cells results in an increase in p53 levels, as well as the expression of the p53-responsive genes p21 and Bax. Furthermore, evidence is provided that increased Cdk5 activity increases p53 transcriptional activity significantly, suggesting that p53 is modulated in situ by Cdk5. This is the first demonstration that p53 is a substrate of Cdk5, and that Cdk5 can modulate p53 levels and activity.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2 , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , CHO Cells , Cell Line , Cricetinae , Cyclin-Dependent Kinase 5 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/pharmacology , Cyclins/metabolism , Enzyme Inhibitors/pharmacology , Genes, Dominant , Humans , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , PC12 Cells , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Purines/pharmacology , Rats , Roscovitine , Substrate Specificity/physiology , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Transfection , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...