Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Physiol Biochem Zool ; 95(5): 390-399, 2022.
Article in English | MEDLINE | ID: mdl-35930827

ABSTRACT

AbstractMammalian hibernation in ground squirrels is characterized by periods of torpor wherein body temperature approaches ambient temperature and metabolism is reduced to as low as 1/100th of active rates. It is unclear how hibernation affects long-term spatial memory, as tremendous remodeling of neurons is associated with torpor use. Given the suspected links between remodeling and memory formation and retention, we examined long-term spatial memory retention throughout a hibernation season. Animals were trained on a Barnes maze before entering torpor. Animals were tested for memory retention once a month throughout a hibernation season. Results indicate marked variation between individuals. Some squirrels retained memory across multiple torpor bouts, while other squirrels did not. No relationship was found between the number of torpor bouts, duration of bouts, or time spent torpid on long-term memory retention. However, that some squirrels successfully retain memory suggests that the profound remodeling of dendritic spines during torpor does not always lead to memory loss.


Subject(s)
Hibernation , Sciuridae , Animals , Body Temperature/physiology , Hibernation/physiology , Sciuridae/physiology , Seasons , Spatial Memory
2.
Cells ; 9(1)2020 01 14.
Article in English | MEDLINE | ID: mdl-31947657

ABSTRACT

Tau dysfunction is common in several neurodegenerative diseases including Alzheimer's disease (AD) and frontotemporal dementia (FTD). Affective symptoms have often been associated with aberrant tau pathology and are commonly comorbid in patients with tauopathies, indicating a connection between tau functioning and mechanisms of depression. The current study investigated depression-like behavior in Mapt-/- mice, which contain a targeted deletion of the gene coding for tau. We show that 6-month Mapt-/- mice are resistant to depressive behaviors, as evidenced by decreased immobility time in the forced swim and tail suspension tests, as well as increased escape behavior in a learned helplessness task. Since depression has also been linked to deficient adult neurogenesis, we measured neurogenesis in the hippocampal dentate gyrus and subventricular zone using 5-bromo-2-deoxyuridine (BrdU) labeling. We found that neurogenesis is increased in the dentate gyrus of 14-month-old Mapt-/- brains compared to wild type, providing a potential mechanism for their behavioral phenotypes. In addition to the hippocampus, an upregulation of proteins involved in neurogenesis was observed in the frontal cortex and amygdala of the Mapt-/- mice using proteomic mass spectrometry. All together, these findings suggest that tau may have a role in the depressive symptoms observed in many neurodegenerative diseases and identify tau as a potential molecular target for treating depression.


Subject(s)
Hippocampus/metabolism , Neurogenesis , Neurons/metabolism , tau Proteins/deficiency , tau Proteins/metabolism , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology
3.
ACS Chem Biol ; 13(8): 2288-2299, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29893552

ABSTRACT

Genetic and epigenetic alterations in FK506-binding protein 5 ( FKBP5) have been associated with increased risk for psychiatric disorders, including post-traumatic stress disorder (PTSD). Some of these common variants can increase the expression of FKBP5, the gene that encodes FKBP51. Excess FKBP51 promotes hypothalamic-pituitary-adrenal (HPA) axis dysregulation through altered glucocorticoid receptor (GR) signaling. Thus, we hypothesized that GR activity could be restored by perturbing FKBP51. Here, we screened 1280 pharmacologically active compounds and identified three compounds that rescued FKBP51-mediated suppression of GR activity without directly activating GR. One of the three compounds, benztropine mesylate, disrupted the association of FKBP51 with the GR/Hsp90 complex in vitro. Moreover, we show that removal of FKBP51 from this complex by benztropine restored GR localization in ex vivo brain slices and primary neurons from mice. In conclusion, we have identified a novel disruptor of the FKBP51/GR/Hsp90 complex. Targeting this complex may be a viable approach to developing treatments for disorders related to aberrant FKBP51 expression.


Subject(s)
Benztropine/pharmacology , Depression/drug therapy , HSP90 Heat-Shock Proteins/metabolism , Receptors, Glucocorticoid/metabolism , Stress Disorders, Post-Traumatic/drug therapy , Tacrolimus Binding Proteins/metabolism , Animals , Benztropine/chemistry , Brain/drug effects , Brain/metabolism , Cells, Cultured , Depression/metabolism , Drug Discovery , Humans , Mice , Molecular Targeted Therapy , Protein Binding/drug effects , Stress Disorders, Post-Traumatic/metabolism , Tacrolimus Binding Proteins/antagonists & inhibitors
4.
Proc Natl Acad Sci U S A ; 114(36): 9707-9712, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28827321

ABSTRACT

The microtubule-associated protein tau (MAPT, tau) forms neurotoxic aggregates that promote cognitive deficits in tauopathies, the most common of which is Alzheimer's disease (AD). The 90-kDa heat shock protein (Hsp90) chaperone system affects the accumulation of these toxic tau species, which can be modulated with Hsp90 inhibitors. However, many Hsp90 inhibitors are not blood-brain barrier-permeable, and several present associated toxicities. Here, we find that the cochaperone, activator of Hsp90 ATPase homolog 1 (Aha1), dramatically increased the production of aggregated tau. Treatment with an Aha1 inhibitor, KU-177, dramatically reduced the accumulation of insoluble tau. Aha1 colocalized with tau pathology in human brain tissue, and this association positively correlated with AD progression. Aha1 overexpression in the rTg4510 tau transgenic mouse model promoted insoluble and oligomeric tau accumulation leading to a physiological deficit in cognitive function. Overall, these data demonstrate that Aha1 contributes to tau fibril formation and neurotoxicity through Hsp90. This suggests that therapeutics targeting Aha1 may reduce toxic tau oligomers and slow or prevent neurodegenerative disease progression.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/pathology , Cell Line , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/genetics , Protein Aggregates , Protein Aggregation, Pathological/etiology , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/prevention & control , Tauopathies/etiology , Tauopathies/metabolism , Tauopathies/prevention & control , tau Proteins/chemistry , tau Proteins/metabolism
5.
PLoS Biol ; 15(6): e2001336, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28654636

ABSTRACT

The accumulation of amyloidogenic proteins is a pathological hallmark of neurodegenerative disorders. The aberrant accumulation of the microtubule associating protein tau (MAPT, tau) into toxic oligomers and amyloid deposits is a primary pathology in tauopathies, the most common of which is Alzheimer's disease (AD). Intrinsically disordered proteins, like tau, are enriched with proline residues that regulate both secondary structure and aggregation propensity. The orientation of proline residues is regulated by cis/trans peptidyl-prolyl isomerases (PPIases). Here we show that cyclophilin 40 (CyP40), a PPIase, dissolves tau amyloids in vitro. Additionally, CyP40 ameliorated silver-positive and oligomeric tau species in a mouse model of tau accumulation, preserving neuronal health and cognition. Nuclear magnetic resonance (NMR) revealed that CyP40 interacts with tau at sites rich in proline residues. CyP40 was also able to interact with and disaggregate other aggregating proteins that contain prolines. Moreover, CyP40 lacking PPIase activity prevented its capacity for disaggregation in vitro. Finally, we describe a unique structural property of CyP40 that may permit disaggregation to occur in an energy-independent manner. This study identifies a novel human protein disaggregase and, for the first time, demonstrates its capacity to dissolve intracellular amyloids.


Subject(s)
Amyloid/metabolism , Cyclophilins/metabolism , Neurodegenerative Diseases/metabolism , alpha-Synuclein/metabolism , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid/genetics , Amyloid/ultrastructure , Animals , Blotting, Western , Brain/metabolism , Brain/pathology , Brain/physiopathology , Cognition Disorders/genetics , Cognition Disorders/metabolism , Cognition Disorders/physiopathology , Peptidyl-Prolyl Isomerase F , Cyclophilins/genetics , Cyclosporine/pharmacology , Disease Models, Animal , Female , HEK293 Cells , Humans , Male , Mice, Transgenic , Microscopy, Electron, Transmission , Neurodegenerative Diseases/genetics , Protein Aggregates/drug effects , Protein Aggregation, Pathological , Tauopathies/genetics , Tauopathies/metabolism , alpha-Synuclein/genetics , tau Proteins/genetics
6.
EMBO J ; 35(14): 1537-49, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27261198

ABSTRACT

It is now known that proteins associated with neurodegenerative disease can spread throughout the brain in a prionlike manner. However, the mechanisms regulating the trans-synaptic spread propagation, including the neuronal release of these proteins, remain unknown. The interaction of neurodegenerative disease-associated proteins with the molecular chaperone Hsc70 is well known, and we hypothesized that much like disaggregation, refolding, degradation, and even normal function, Hsc70 may dictate the extracellular fate of these proteins. Here, we show that several proteins, including TDP-43, α-synuclein, and the microtubule-associated protein tau, can be driven out of the cell by an Hsc70 co-chaperone, DnaJC5. In fact, DnaJC5 overexpression induced tau release in cells, neurons, and brain tissue, but only when activity of the chaperone Hsc70 was intact and when tau was able to associate with this chaperone. Moreover, release of tau from neurons was reduced in mice lacking the DnaJC5 gene and when the complement of DnaJs in the cell was altered. These results demonstrate that the dynamics of DnaJ/Hsc70 complexes are critically involved in the release of neurodegenerative disease proteins.


Subject(s)
HSC70 Heat-Shock Proteins/metabolism , HSP40 Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , tau Proteins/metabolism , Cell Line , DNA-Binding Proteins/metabolism , Humans , alpha-Synuclein/metabolism
7.
J Biol Chem ; 291(34): 17897-906, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27334923

ABSTRACT

Single nucleotide polymorphisms in the FKBP5 gene increase the expression of the FKBP51 protein and have been associated with increased risk for neuropsychiatric disorders such as major depression and post-traumatic stress disorder. Moreover, levels of FKBP51 are increased with aging and in Alzheimer disease, potentially contributing to disease pathogenesis. However, aside from its glucocorticoid responsiveness, little is known about what regulates FKBP5 In recent years, non-coding RNAs, and in particular microRNAs, have been shown to modulate disease-related genes and processes. The current study sought to investigate which miRNAs could target and functionally regulate FKBP5 Following in silico data mining and initial target expression validation, miR-511 was found to suppress FKBP5 mRNA and protein levels. Using luciferase p-miR-Report constructs and RNA pulldown assays, we confirmed that miR-511 bound directly to the 3'-UTR of FKBP5, validating the predicted gene-microRNA interaction. miR-511 suppressed glucocorticoid-induced up-regulation of FKBP51 in cells and primary neurons, demonstrating functional, disease-relevant control of the protein. Consistent with a regulator of FKBP5, miR-511 expression in the mouse brain decreased with age but increased following chronic glucocorticoid treatment. Analysis of the predicted target genes of miR-511 revealed that neurogenesis, neuronal development, and neuronal differentiation were likely controlled by these genes. Accordingly, miR-511 increased neuronal differentiation in cells and enhanced neuronal development in primary neurons. Collectively, these findings show that miR-511 is a functional regulator of FKBP5 and can contribute to neuronal differentiation.


Subject(s)
3' Untranslated Regions/physiology , Brain/metabolism , Cell Differentiation/physiology , Gene Expression Regulation/physiology , MicroRNAs/metabolism , Molecular Chaperones/biosynthesis , Neurogenesis/physiology , Neurons/metabolism , Tacrolimus Binding Proteins/biosynthesis , Animals , HeLa Cells , Humans , Mice , MicroRNAs/genetics , Molecular Chaperones/genetics , Tacrolimus Binding Proteins/genetics
8.
ACS Chem Biol ; 11(7): 2041-8, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27177119

ABSTRACT

Three scaffolds with inhibitory activity against the heat shock protein 70 (Hsp70) family of chaperones have been found to enhance the degradation of the microtubule associated protein tau in cells, neurons, and brain tissue. This is important because tau accumulation is linked to neurodegenerative diseases including Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). Here, we expanded upon this study to investigate the anti-tau efficacy of additional scaffolds with Hsp70 inhibitory activity. Five of the nine scaffolds tested lowered tau levels, with the rhodacyanine and phenothiazine scaffolds exhibiting the highest potency as previously described. Because phenothiazines also inhibit tau aggregation in vitro, we suspected that this activity might be a more accurate predictor of tau lowering. Interestingly, the rhodacyanines did inhibit in vitro tau aggregation to a similar degree as phenothiazines, correlating well with tau-lowering efficacy in cells and ex vivo slices. Moreover, other Hsp70 inhibitor scaffolds with weaker tau-lowering activity in cells inhibited tau aggregation in vitro, albeit at lower potencies. When we tested six well-characterized tau aggregation inhibitors, we determined that this mechanism of action was not a better predictor of tau-lowering than Hsp70 inhibition. Instead, we found that compounds possessing both activities were the most effective at promoting tau clearance. Moreover, cytotoxicity and PAINS activity are critical factors that can lead to false-positive lead identification. Strategies designed around these principles will likely yield more efficacious tau-lowering compounds.


Subject(s)
HSP70 Heat-Shock Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , tau Proteins/antagonists & inhibitors , HEK293 Cells , Humans , In Vitro Techniques
9.
Front Neurosci ; 10: 3, 2016.
Article in English | MEDLINE | ID: mdl-26834532

ABSTRACT

Accumulation of the microtubule associated protein tau occurs in several neurodegenerative diseases including Alzheimer's disease (AD). The tau protein is intrinsically disordered, giving it unique structural properties that can be dynamically altered by post-translational modifications such as phosphorylation and cleavage. Over the last decade, technological advances in nuclear magnetic resonance (NMR) spectroscopy and structural modeling have permitted more in-depth insights into the nature of tau. These studies have helped elucidate how metamorphism of tau makes it ideally suited for dynamic microtubule regulation, but how it also facilitates tau self-assembly, oligomerization, and neurotoxicity. This review will focus on how the distinct structure of tau governs its function, accumulation, and toxicity as well as how other cellular factors such as molecular chaperones control these processes.

10.
J Neurotrauma ; 33(19): 1751-1760, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-26739819

ABSTRACT

Traumatic brain injury (TBI) caused by improvised explosive devices (IEDs) is a growing problem in military settings, but modeling this disease in rodents to pre-clinically evaluate potential therapeutics has been challenging because of inconsistency between models. Although the effects of primary blast wave injury have been extensively studied, little is known regarding the effects of noncontact rotational TBIs independent of the blast wave. To model this type of injury, we generated an air cannon system that does not produce a blast wave, but generates enough air pressure to cause rotational TBI. Mice exposed to this type of injury showed deficits in cognitive and motor task acquisition within 1-2 weeks post-injury, but mice tested 7-8 weeks post-injury did not retain any deficits. This suggests that the effects of a single, noncontact rotational TBI are not long lasting. Despite the transient nature of the behavioral deficits, increased levels of phosphorylated tau were observed at 2 and 8 weeks post-injury; however, this tau did not adopt typical pathological structures that have been observed in other TBI models that incorporate blast waves. This was possibly attributed to the fact that this injury was insufficient to induce changes in microglial activation, which was not affected at 2 or 8 weeks post-injury. Taken together, these data suggest that exposure to noncontact, rotational head injury only produces transient cognitive anomalies, but elicits some minor lasting neuropathological changes.

11.
Neuropharmacology ; 101: 110-22, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26327677

ABSTRACT

Lipopolysaccharide (LPS) is often used to investigate the exacerbatory effects of an immune-related challenge in transgenic models of various neurodegenerative diseases. However, the effects of this inflammatory challenge in an insulin resistant brain state, as seen in diabetes mellitus, a major risk factor for both vascular dementia (VaD) and Alzheimer's disease (AD), is not as well characterized. We investigated the effects of an LPS-induced inflammatory challenge on behavioral and biological parameters following intracerebroventricular (ICV) injection of streptozotocin (STZ) in male Sprague-Dawley rats. Subjects received a one-time bilateral ICV infusion of STZ (25 mg/mL, 8 µL per ventricle) or ACSF. One week following ICV infusions, LPS (1 mg/mL, i.p.) or saline was administered to activate the immune system. Behavioral testing began on the 22nd day following STZ-ICV infusion, utilizing the open field and Morris water maze (MWM) tasks. Proteins related to immune function, learning and memory, synaptic plasticity, and key histopathological markers observed in VaD and AD were evaluated. The addition of an LPS-induced immune challenge partially attenuated spatial learning and memory deficits in the MWM in STZ-ICV injected animals. Additionally, LPS administration to STZ-treated animals partially mitigated alterations observed in several protein levels in STZ-ICV alone, including NR2A, GABA(B1), and ß-amyloid oligomers. These results suggest that an acute LPS-inflammatory response has a modest protective effect against some of the spatial learning and memory deficits and protein alterations associated with STZ-ICV induction of an insulin resistant brain state.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Inflammation/chemically induced , Lipopolysaccharides/toxicity , Streptozocin/administration & dosage , Amyloid beta-Peptides/metabolism , Analysis of Variance , Animals , Disease Models, Animal , Exploratory Behavior/drug effects , Guanine Nucleotide Exchange Factors/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Inflammation/metabolism , Inflammation/physiopathology , Injections, Intraventricular , Interleukin-6/metabolism , Male , Maze Learning/drug effects , Rats , Rats, Sprague-Dawley , Receptors, GABA-B/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Time Factors
12.
Exp Eye Res ; 144: 38-45, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26302411

ABSTRACT

A major drainage network involved in aqueous humor dynamics is the conventional outflow pathway, which is gated by the trabecular meshwork (TM). The TM acts as a molecular sieve, providing resistance to aqueous outflow, which is responsible for regulating intraocular pressure (IOP). If the TM is damaged, aqueous outflow is impaired, IOP increases and glaucoma can manifest. Mutations in the MYOC gene cause hereditary primary open-angle glaucoma (POAG) by promoting the abnormal amyloidosis of the myocilin protein in the endoplasmic reticulum (ER), leading to ER stress-induced TM cell death. Myocilin accumulation is observed in approximately 70-80% of all glaucoma cases suggesting that environmental or other genetic factors may also promote myocilin toxicity. For example, simply preventing myocilin glycosylation is sufficient to promote its abnormal accretion. These myocilin amyloids are unique as there are no other known pathogenic proteins that accumulate within the ER of TM cells and cause toxicity. Moreover, this pathogenic accumulation only kills TM cells, despite expression of this protein in other cell types, suggesting that another modifier exclusive to the TM participates in the proteotoxicity of myocilin. ER autophagy (reticulophagy) is one of the pathways essential for myocilin clearance that can be impacted dramatically by aging and other environmental factors such as nutrition. This review will discuss the link between myocilin and autophagy, evaluating the role of this degradation pathway in glaucoma as well as its potential as a therapeutic target.


Subject(s)
Autophagy/drug effects , Endoplasmic Reticulum/drug effects , Glaucoma, Open-Angle/drug therapy , Molecular Targeted Therapy , Animals , Aqueous Humor/physiology , Cytoskeletal Proteins/metabolism , Eye Proteins/metabolism , Glaucoma, Open-Angle/metabolism , Glycoproteins/metabolism , Humans , Intraocular Pressure/physiology , Trabecular Meshwork
13.
Hum Mol Genet ; 24(14): 3971-81, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25882706

ABSTRACT

The pathological accumulation of abnormally hyperphosphorylated and aggregated tau, a neuronal microtubule (MT)-associated protein that functions to maintain MT stability, is implicated in a number of hereditary and sporadic neurodegenerative diseases including frontotemporal dementia and Alzheimer's disease. Targeting tau for the treatment of these diseases is an area of intense interest and toward that end, modulation of cellular molecular chaperones is a potential therapeutic target. In particular, the constitutive Hsp70 isoform, Hsc70, seems highly interconnected with tau, preserving tau protein levels and synergizing with it to assemble MTs. But the relationship between tau and Hsc70, as well as the impact of this interaction in neurons and its therapeutic implications remain unknown. Using a human dominant negative Hsc70 that resembles isoform selective inhibition of this important chaperone, we found for the first time that Hsc70 activity is required to stimulate MT assembly in cells and brain. However, surprisingly, active Hsc70 also requires active tau to regulate MT assembly in vivo, suggesting that tau acts in some ways as a co-chaperone for Hsc70 to coordinate MT assembly. This was despite tau binding to Hsc70 as substrate, as determined biochemically. Moreover, we show that while chronic Hsc70 inhibition damaged MT dynamics, intermittent treatment with a small molecule Hsp70 inhibitor lowered tau in brain tissue without disrupting MT integrity. Thus, in tauopathies, where MT injury would be detrimental to neurons, the unique relationship of tau with the Hsc70 machinery can be exploited to deplete tau levels without damaging MT networks.


Subject(s)
HSC70 Heat-Shock Proteins/metabolism , Microtubules/metabolism , tau Proteins/metabolism , Animals , Brain/metabolism , Gene Expression Regulation , HEK293 Cells , HSC70 Heat-Shock Proteins/genetics , Humans , Magnetic Resonance Spectroscopy , Mice, Knockout , Neurons/metabolism , Oocytes , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tauopathies/genetics , Tauopathies/therapy , Xenopus , tau Proteins/genetics
14.
J Biol Chem ; 290(21): 13115-27, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25864199

ABSTRACT

The constitutively expressed heat shock protein 70 kDa (Hsc70) is a major chaperone protein responsible for maintaining proteostasis, yet how its structure translates into functional decisions regarding client fate is still unclear. We previously showed that Hsc70 preserved aberrant Tau, but it remained unknown if selective inhibition of the activity of this Hsp70 isoform could facilitate Tau clearance. Using single point mutations in the nucleotide binding domain, we assessed the effect of several mutations on the functions of human Hsc70. Biochemical characterization revealed that one mutation abolished both Hsc70 ATPase and refolding activities. This variant resembled the ADP-bound conformer at all times yet remained able to interact with cofactors, nucleotides, and substrates appropriately, resembling a dominant negative Hsc70 (DN-Hsc70). We then assessed the effects of this DN-Hsc70 on its client Tau. DN-Hsc70 potently facilitated Tau clearance via the proteasome in cells and brain tissue, in contrast to wild type Hsc70 that stabilized Tau. Thus, DN-Hsc70 mimics the action of small molecule pan Hsp70 inhibitors with regard to Tau metabolism. This shift in Hsc70 function by a single point mutation was the result of a change in the chaperome associated with Hsc70 such that DN-Hsc70 associated more with Hsp90 and DnaJ proteins, whereas wild type Hsc70 was more associated with other Hsp70 isoforms. Thus, isoform-selective targeting of Hsc70 could be a viable therapeutic strategy for tauopathies and possibly lead to new insights in chaperone complex biology.


Subject(s)
Adenosine Triphosphatases/metabolism , HSC70 Heat-Shock Proteins/antagonists & inhibitors , HSC70 Heat-Shock Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Folding , tau Proteins/metabolism , Blotting, Western , Cells, Cultured , Cytosol/metabolism , Fluorescence Polarization , Fluorescent Antibody Technique , HSC70 Heat-Shock Proteins/genetics , Humans , Magnetic Resonance Spectroscopy , Mutation/genetics , Protein Binding , Protein Conformation , Protein Isoforms , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , tau Proteins/genetics
15.
Acta Neuropathol Commun ; 3: 8, 2015 Jan 31.
Article in English | MEDLINE | ID: mdl-25775028

ABSTRACT

INTRODUCTION: The blood-brain barrier (BBB) is damaged in tauopathies, including progressive supranuclear palsy (PSP) and Alzheimer's disease (AD), which is thought to contribute to pathogenesis later in the disease course. In AD, BBB dysfunction has been associated with amyloid beta (Aß) pathology, but the role of tau in this process is not well characterized. Since increased BBB permeability is found in tauopathies without Aß pathology, like PSP, we suspected that tau accumulation alone could not only be sufficient, but even more important than Aß for BBB damage. RESULTS: Longitudinal evaluation of brain tissue from the tetracycline-regulatable rTg4510 tau transgenic mouse model showed progressive IgG, T cell and red blood cell infiltration. The Evans blue (EB) dye that is excluded from the brain when the BBB is intact also permeated the brains of rTg4510 mice following peripheral administration, indicative of a bonafide BBB defect, but this was only evident later in life. Thus, despite the marked brain atrophy and inflammation that occurs earlier in this model, BBB integrity is maintained. Interestingly, BBB dysfunction emerged at the same time that perivascular tau emerged around major hippocampal blood vessels. However, when tau expression was suppressed using doxycycline, BBB integrity was preserved, suggesting that the BBB can be stabilized in a tauopathic brain by reducing tau levels. CONCLUSIONS: For the first time, these data demonstrate that tau alone can initiate breakdown of the BBB, but the BBB is remarkably resilient, maintaining its integrity in the face of marked brain atrophy, neuroinflammation and toxic tau accumulation. Moreover, the BBB can recover integrity when tau levels are reduced. Thus, late stage interventions targeting tau may slow the vascular contributions to cognitive impairment and dementia that occur in tauopathies.


Subject(s)
Blood-Brain Barrier/physiopathology , Gene Expression Regulation/genetics , Tauopathies/pathology , tau Proteins/deficiency , Age Factors , Analysis of Variance , Animals , Antigens, CD/metabolism , Blood-Brain Barrier/drug effects , Disease Models, Animal , Disease Progression , Doxycycline/pharmacology , Erythrocytes/pathology , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , HSP27 Heat-Shock Proteins/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Immunoglobulin G/metabolism , Longitudinal Studies , Mice , Mice, Transgenic , T-Lymphocytes/pathology , tau Proteins/genetics
16.
Cell Mol Life Sci ; 72(10): 1863-79, 2015 May.
Article in English | MEDLINE | ID: mdl-25666877

ABSTRACT

Pathological accumulation of the microtubule-associated protein tau, in the form of neurofibrillary tangles, is a major hallmark of Alzheimer's disease, the most prevalent neurodegenerative condition worldwide. In addition to Alzheimer's disease, a number of neurodegenerative diseases, called tauopathies, are characterized by the accumulation of aggregated tau in a variety of brain regions. While tau normally plays an important role in stabilizing the microtubule network of the cytoskeleton, its dissociation from microtubules and eventual aggregation into pathological deposits is an area of intense focus for therapeutic development. Here we discuss the known cellular factors that affect tau aggregation, from post-translational modifications to molecular chaperones.


Subject(s)
Alzheimer Disease/physiopathology , Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Protein Aggregation, Pathological/metabolism , tau Proteins/metabolism , Acetylation , Glycosylation , Humans , Microtubules/metabolism , Phosphorylation , Proteolysis , tau Proteins/genetics
17.
J Neurochem ; 133(1): 1-13, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25628064

ABSTRACT

Peptidyl-prolyl cis/trans isomerases (PPIases), a unique family of molecular chaperones, regulate protein folding at proline residues. These residues are abundant within intrinsically disordered proteins, like the microtubule-associated protein tau. Tau has been shown to become hyperphosphorylated and accumulate as one of the two main pathological hallmarks in Alzheimer's disease, the other being amyloid beta (Ab). PPIases, including Pin1, FK506-binding protein (FKBP) 52, FKBP51, and FKBP12, have been shown to interact with and regulate tau biology. This interaction is particularly important given the numerous proline-directed phosphorylation sites found on tau and the role phosphorylation has been found to play in pathogenesis. This regulation then affects downstream aggregation and oligomerization of tau. However, many PPIases have yet to be explored for their effects on tau biology, despite the high likelihood of interaction based on proline content. Moreover, Pin1, FKBP12, FKBP52, cyclophilin (Cyp) A, CypB, and CypD have been shown to also regulate Ab production or the toxicity associated with Ab pathology. Therefore, PPIases directly and indirectly regulate pathogenic protein multimerization in Alzheimer's disease and represent a family rich in targets for modulating the accumulation and toxicity.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Molecular Chaperones/metabolism , Peptidylprolyl Isomerase/metabolism , tau Proteins/metabolism , Animals , Humans
18.
Int J Dev Neurosci ; 41: 17-27, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25314921

ABSTRACT

The GABA transmitter system plays a vital role in modulating synaptic formation and activity during development. The GABAB receptor subtype in particular has been implicated in cell migration, promotion of neuronal differentiation, neurite outgrowth, and synapse formation but it's role in development is not well characterized. In order to investigate the effects of brief alterations in GABAB signaling in development, we administered to rats the GABAB agonist baclofen (2.0mg/kg) or antagonist phaclofen (0.3mg/kg) on postnatal days 7, 9, and 12, and evaluated sensorimotor gating in adulthood. We also examined tissue for changes in multiple proteins associated with GABAB receptor function and proteins associated with synapse formation. Our data indicate that early postnatal alterations to GABAB receptor-mediated signaling produced sex differences in sensorimotor gating in adulthood. Additionally, we found differences in GABAB receptor subunits and kalirin protein levels in the brain versus saline treated controls. Our data demonstrate that a subtle alteration in GABAB receptor function in early postnatal life induces changes that persist into adulthood.


Subject(s)
Brain/metabolism , Gait Disorders, Neurologic/metabolism , Gait Disorders, Neurologic/pathology , Gene Expression Regulation, Developmental/physiology , Receptors, GABA-B/metabolism , Signal Transduction/physiology , Acoustic Stimulation , Age Factors , Animals , Animals, Newborn , Baclofen/analogs & derivatives , Baclofen/toxicity , Body Weight/drug effects , Disease Models, Animal , Female , GABA Antagonists/toxicity , GABA-B Receptor Agonists/toxicity , Gait Disorders, Neurologic/chemically induced , Gene Expression Regulation, Developmental/drug effects , Male , Pregnancy , Prepulse Inhibition/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley , Sensory Gating/drug effects , Signal Transduction/drug effects
19.
PLoS One ; 9(9): e107241, 2014.
Article in English | MEDLINE | ID: mdl-25191701

ABSTRACT

Single nucleotide polymorphisms (SNPs) in the FK506 binding protein 5 (FKBP5) gene combine with traumatic events to increase risk for post-traumatic stress and major depressive disorders (PTSD and MDD). These SNPs increase FKBP51 protein expression through a mechanism involving demethylation of the gene and altered glucocorticoid signaling. Aged animals also display elevated FKBP51 levels, which contribute to impaired resiliency to depressive-like behaviors through impaired glucocorticoid signaling, a phenotype that is abrogated in FKBP5-/- mice. But the age of onset and progressive stability of these phenotypes remain unknown. Moreover, it is unclear how FKBP5 deletion affects other glucocorticoid-dependent processes or if age-associated increases in FKBP51 expression are mediated through a similar epigenetic process caused by SNPs in the FKBP5 gene. Here, we show that FKBP51-mediated impairment in stress resiliency and glucocorticoid signaling occurs by 10 months of age and this increased over their lifespan. Surprisingly, despite these progressive changes in glucocorticoid responsiveness, FKBP5-/- mice displayed normal longevity, glucose tolerance, blood composition and cytokine profiles across lifespan, phenotypes normally associated with glucocorticoid signaling. We also found that methylation of Fkbp5 decreased with age in mice, a process that likely explains the age-associated increases in FKBP51 levels. Thus, epigenetic upregulation of FKBP51 with age can selectively impair psychological stress-resiliency, but does not affect other glucocorticoid-mediated physiological processes. This makes FKBP51 a unique and attractive therapeutic target to treat PTSD and MDD. In addition, aged wild-type mice may be a useful model for investigating the mechanisms of FKBP5 SNPs associated with these disorders.


Subject(s)
Aging/genetics , Epigenesis, Genetic/physiology , Resilience, Psychological , Stress, Psychological/genetics , Tacrolimus Binding Proteins/genetics , Aging/blood , Animals , DNA Methylation , Depressive Disorder, Major/genetics , Hydrocortisone/blood , Longevity/genetics , Mice , Mice, Knockout , Polymorphism, Single Nucleotide , Stress Disorders, Post-Traumatic/genetics , Stress, Psychological/blood , Tacrolimus Binding Proteins/metabolism , Up-Regulation/genetics
20.
Expert Opin Ther Targets ; 18(10): 1219-32, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25069659

ABSTRACT

INTRODUCTION: Alzheimer's disease, characterized by the accumulation of hyperphosphorylated tau and ß amyloid (Aß), currently lacks effective treatment. Chaperone proteins, such as the heat shock protein (Hsp) 90, form macromolecular complexes with co-chaperones, which can regulate tau metabolism and Aß processing. Although small molecule inhibitors of Hsp90 have been successful at ameliorating tau and Aß burden, their development into drugs to treat disease has been slow due to the off- and on-target effects of this approach as well as challenges with the pharmacology of current scaffolds. Thus, other approaches are being developed to improve these compounds and to target co-chaperones of Hsp90 in an effort to limit these liabilities. AREAS COVERED: This article discusses the most current developments in Hsp90 inhibitors including advances in blood-brain barrier permeability, decreased toxicity and homolog-specific small-molecule inhibitors. In addition, we discuss current strategies targeting Hsp90 co-chaperones rather than Hsp90 itself to reduce off-target effects. EXPERT OPINION: Although Hsp90 inhibitors have proven their efficacy at reducing tau pathology, they have yet to meet with success in the clinic. The development of Hsp90/tau complex-specific inhibitors and further development of Hsp90 co-chaperone-specific drugs should yield more potent, less toxic therapeutics.


Subject(s)
Alzheimer Disease/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Molecular Targeted Therapy , Alzheimer Disease/physiopathology , Animals , Blood-Brain Barrier/metabolism , Drug Design , HSP90 Heat-Shock Proteins/metabolism , Humans , Molecular Chaperones/metabolism , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...