Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 5149, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890299

ABSTRACT

Telomeres are the protective nucleoprotein structures at the end of linear eukaryotic chromosomes. Telomeres' repetitive nature and length have traditionally challenged the precise assessment of the composition and length of individual human telomeres. Here, we present Telo-seq to resolve bulk, chromosome arm-specific and allele-specific human telomere lengths using Oxford Nanopore Technologies' native long-read sequencing. Telo-seq resolves telomere shortening in five population doubling increments and reveals intrasample, chromosome arm-specific, allele-specific telomere length heterogeneity. Telo-seq can reliably discriminate between telomerase- and ALT-positive cancer cell lines. Thus, Telo-seq is a tool to study telomere biology during development, aging, and cancer at unprecedented resolution.


Subject(s)
Aging , Neoplasms , Telomere , Humans , Telomere/genetics , Telomere/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Aging/genetics , Telomerase/genetics , Telomerase/metabolism , Cell Line, Tumor , Telomere Shortening/genetics , Sequence Analysis, DNA/methods , High-Throughput Nucleotide Sequencing/methods , Alleles
3.
Cell Rep ; 42(12): 113466, 2023 12 26.
Article in English | MEDLINE | ID: mdl-38039131

ABSTRACT

Biallelic mutations in the gene that encodes the enzyme N-glycanase 1 (NGLY1) cause a rare disease with multi-symptomatic features including developmental delay, intellectual disability, neuropathy, and seizures. NGLY1's activity in human neural cells is currently not well understood. To understand how NGLY1 gene loss leads to the specific phenotypes of NGLY1 deficiency, we employed direct conversion of NGLY1 patient-derived induced pluripotent stem cells (iPSCs) to functional cortical neurons. Transcriptomic, proteomic, and functional studies of iPSC-derived neurons lacking NGLY1 function revealed several major cellular processes that were altered, including protein aggregate-clearing functionality, mitochondrial homeostasis, and synaptic dysfunctions. These phenotypes were rescued by introduction of a functional NGLY1 gene and were observed in iPSC-derived mature neurons but not astrocytes. Finally, laser capture microscopy followed by mass spectrometry provided detailed characterization of the composition of protein aggregates specific to NGLY1-deficient neurons. Future studies will harness this knowledge for therapeutic development.


Subject(s)
Protein Aggregates , Proteomics , Humans , Mutation/genetics , Mitochondria/metabolism , Neurons/metabolism , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase
4.
Nat Rev Neurol ; 19(7): 434-443, 2023 07.
Article in English | MEDLINE | ID: mdl-37268723

ABSTRACT

Most age-related neurodegenerative diseases remain incurable owing to an incomplete understanding of the disease mechanisms. Several environmental and genetic factors contribute to disease onset, with human biological ageing being the primary risk factor. In response to acute cellular damage and external stimuli, somatic cells undergo state shifts characterized by temporal changes in their structure and function that increase their resilience, repair cellular damage, and lead to their mobilization to counteract the pathology. This basic cell biological principle also applies to human brain cells, including mature neurons that upregulate developmental features such as cell cycle markers or glycolytic reprogramming in response to stress. Although such temporary state shifts are required to sustain the function and resilience of the young human brain, excessive state shifts in the aged brain might result in terminal fate loss of neurons and glia, characterized by a permanent change in cell identity. Here, we offer a new perspective on the roles of cell states in sustaining health and counteracting disease, and we examine how cellular ageing might set the stage for pathological fate loss and neurodegeneration. A better understanding of neuronal state and fate shifts might provide the means for a controlled manipulation of cell fate to promote brain resilience and repair.


Subject(s)
Neurodegenerative Diseases , Neurons , Humans , Aged , Neurons/physiology , Neuroglia/metabolism , Brain , Neurodegenerative Diseases/metabolism , Aging
5.
J Dent Educ ; 87(9): 1250-1256, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37208799

ABSTRACT

PURPOSE/OBJECTIVES: Developing time management is an important aspect of a dental student's passage to clinical care and in their growth as a professional. Suitable time management and preparedness can potentially impact the prognosis of a successful dental appointment. The objective of this study was to determine if a time management exercise could be effective to increase students' preparedness, organization, time management, and reflection during simulated clinical care prior to transitioning to the dental clinic. METHODS: Students completed five-time management exercises during the term preceding their entrance into the predoctoral restorative clinic which included appointment planning and organization, and reflection once finished. Pre- and post-term surveys were used to determine the impact of the experience. Quantitative data was analyzed using a paired t-test and the qualitative data was thematically coded by the researchers. RESULTS: Students reported a statistically significant increase in their self-confidence of clinical readiness after completion of the time management series, and all students completed the surveys. The themes indicated by students through their comments in the post-survey question regarding the experience were as follows: planning and preparation, time management, the practice of procedures, concern about workload, faculty encouragement, and unclear. Most students also reported the exercise to be beneficial for their predoctoral clinical appointments. CONCLUSIONS: It was determined that the time management exercises were effective for the students as they transitioned to treating patients in the predoctoral clinic and can be used for future classes to increase their success.


Subject(s)
Students, Dental , Time Management , Humans , Self Concept
6.
Science ; 372(6537): 91-94, 2021 04 02.
Article in English | MEDLINE | ID: mdl-33795458

ABSTRACT

Neurons are the longest-lived cells in our bodies and lack DNA replication, which makes them reliant on a limited repertoire of DNA repair mechanisms to maintain genome fidelity. These repair mechanisms decline with age, but we have limited knowledge of how genome instability emerges and what strategies neurons and other long-lived cells may have evolved to protect their genomes over the human life span. A targeted sequencing approach in human embryonic stem cell-induced neurons shows that, in neurons, DNA repair is enriched at well-defined hotspots that protect essential genes. These hotspots are enriched with histone H2A isoforms and RNA binding proteins and are associated with evolutionarily conserved elements of the human genome. These findings provide a basis for understanding genome integrity as it relates to aging and disease in the nervous system.


Subject(s)
DNA Repair , Genome, Human , Genomic Instability , Neurons/metabolism , Aging/genetics , DNA Damage , DNA, Intergenic , Deoxyuridine/analogs & derivatives , Deoxyuridine/metabolism , Embryonic Stem Cells , Histones/metabolism , Humans , Mitosis , Mutation , Nervous System Diseases/genetics , Neurons/cytology , Promoter Regions, Genetic , RNA-Binding Proteins/metabolism , Sequence Analysis, DNA , Transcription, Genetic
7.
Article in English | MEDLINE | ID: mdl-31767646

ABSTRACT

Brain disorders, from neurodegenerative to psychiatric disorders, are among the most challenging conditions to study because of the intricate nature of the human brain and the limitations of existing model systems in recapitulating all these intricacies. However, innovations in stem cell technologies now allow us to reprogram patient somatic cells to induced pluripotent stem cells (iPSCs), which can then be differentiated to disease-relevant neural and glial cells. iPSCs are a valuable tool to model brain disorders, as they can be derived from patients with known symptom histories, genetics, and drug-response profiles. Here, we discuss the premise and validity of the iPSC-based in vitro model system and highlight key findings from the most commonly studied neurodegenerative and psychiatric disorders.


Subject(s)
Brain Diseases/metabolism , Brain/physiopathology , Induced Pluripotent Stem Cells/metabolism , Animals , Brain/physiology , Cell Differentiation , Central Nervous System , Chemistry, Pharmaceutical/methods , Drug Design , Genetic Techniques , Humans , Models, Biological , Models, Neurological , Neurodegenerative Diseases , Neuroglia/metabolism , Neurons/metabolism , Pluripotent Stem Cells/physiology , Stem Cell Transplantation
8.
Cell Rep ; 25(4): 947-958.e4, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30355500

ABSTRACT

How mutations in glial fibrillary acidic protein (GFAP) cause Alexander disease (AxD) remains elusive. We generated iPSCs from two AxD patients and corrected the GFAP mutations to examine the effects of mutant GFAP on human astrocytes. AxD astrocytes displayed GFAP aggregates, recapitulating the pathological hallmark of AxD. RNA sequencing implicated the endoplasmic reticulum, vesicle regulation, and cellular metabolism. Corroborating this analysis, we observed enlarged and heterogeneous morphology coupled with perinuclear localization of endoplasmic reticulum and lysosomes in AxD astrocytes. Functionally, AxD astrocytes showed impaired extracellular ATP release, which is responsible for attenuated calcium wave propagation. These results reveal that AxD-causing mutations in GFAP disrupt intracellular vesicle regulation and impair astrocyte secretion, resulting in astrocyte dysfunction and AxD pathogenesis.


Subject(s)
Astrocytes/metabolism , Glial Fibrillary Acidic Protein/genetics , Mutation/genetics , Organelles/metabolism , Adenosine Triphosphate/metabolism , Alexander Disease/metabolism , Alexander Disease/pathology , Animals , Astrocytes/cytology , Calcium Signaling , Cell Differentiation , Endoplasmic Reticulum/metabolism , Humans , Lysosomes/metabolism , Mice , Protein Aggregates , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
Nat Commun ; 9(1): 1899, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29765022

ABSTRACT

Glial cells have increasingly been implicated as active participants in the pathogenesis of neurological diseases, but critical pathways and mechanisms controlling glial function and secondary non-cell autonomous neuronal injury remain incompletely defined. Here we use models of Alexander disease, a severe brain disorder caused by gain-of-function mutations in GFAP, to demonstrate that misregulation of GFAP leads to activation of a mechanosensitive signaling cascade characterized by activation of the Hippo pathway and consequent increased expression of A-type lamin. Importantly, we use genetics to verify a functional role for dysregulated mechanotransduction signaling in promoting behavioral abnormalities and non-cell autonomous neurodegeneration. Further, we take cell biological and biophysical approaches to suggest that brain tissue stiffness is increased in Alexander disease. Our findings implicate altered mechanotransduction signaling as a key pathological cascade driving neuronal dysfunction and neurodegeneration in Alexander disease, and possibly also in other brain disorders characterized by gliosis.


Subject(s)
Alexander Disease/metabolism , Mechanotransduction, Cellular , Adolescent , Adult , Alexander Disease/genetics , Alexander Disease/psychology , Animals , Behavior, Animal , Biomechanical Phenomena , Brain/metabolism , Brain Chemistry , Child , Child, Preschool , Drosophila , Female , Glial Fibrillary Acidic Protein/metabolism , Hippo Signaling Pathway , Humans , Infant , Lamin Type A/genetics , Lamin Type A/metabolism , Male , Mice , Mice, Transgenic , Neuroglia/chemistry , Neuroglia/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Young Adult
10.
Sci Rep ; 6: 32600, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27600186

ABSTRACT

The process of neuroepithelial differentiation from human pluripotent stem cells (PSCs) resembles in vivo neuroectoderm induction in the temporal course, morphogenesis, and biochemical changes. This in vitro model is therefore well-suited to reveal previously unknown molecular mechanisms underlying neural induction in humans. By transcriptome analysis of cells along PSC differentiation to early neuroepithelia at day 6 and definitive neuroepithelia at day 10, we found downregulation of genes that are associated with TGF-ß and canonical WNT/ß-CATENIN signaling, confirming the roles of classical signaling in human neural induction. Interestingly, WNT/Ca(2+) signaling was upregulated. Pharmacological inhibition of the downstream effector of WNT/Ca(2+) pathway, Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), led to an inhibition of the neural marker PAX6 and upregulation of epidermal marker K18, suggesting that Ca(2+)/CaMKII signaling promotes neural induction by preventing the alternative epidermal fate. In addition, our analyses revealed known and novel expression patterns of genes that are involved in DNA methylation, histone modification, as well as epithelial-mesenchymal transition, highlighting potential roles of those genes and signaling pathways during neural differentiation.


Subject(s)
Gene Expression Profiling , Neuroepithelial Cells/metabolism , Wnt Signaling Pathway/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Differentiation/genetics , Ectoderm/embryology , Ectoderm/metabolism , Epigenesis, Genetic , Epithelial-Mesenchymal Transition/genetics , Extracellular Space/metabolism , Gene Expression Regulation, Developmental , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Neuroepithelial Cells/cytology , Neurogenesis/genetics , Oligonucleotide Array Sequence Analysis , Phosphorylation , Prosencephalon/embryology , Prosencephalon/metabolism , Signal Transduction/genetics , Time Factors
11.
Curr Opin Biotechnol ; 40: 133-138, 2016 08.
Article in English | MEDLINE | ID: mdl-27082135

ABSTRACT

The utility of human pluripotent stem cells (hPSCs) depends on their ability to produce functional cells and tissues of the body. Two strategies have been developed: directed differentiation of enriched populations of cells that match a regional and functional profile and spontaneous generation of three-dimensional organoids that resemble tissues in the body. Genomic editing of hPSCs and their differentiated cells broadens the use of the hPSC paradigm in studying human cellular function and disease as well as developing therapeutics.


Subject(s)
Cell Differentiation , Pluripotent Stem Cells/cytology , RNA Editing/genetics , Tissue Engineering/methods , Humans
12.
Circ Res ; 114(9): 1435-45, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24637197

ABSTRACT

RATIONALE: Myocardial infarction (MI) causes an imbalance between matrix metalloproteinases and tissue inhibitors of matrix metalloproteinases (TIMPs) and is associated with adverse left ventricular (LV) remodeling. A uniform reduction in TIMP-4 post-MI has been observed. OBJECTIVE: To examine post-MI remodeling with cardiac-restricted overexpression of TIMP-4, either through a transgenic or viral delivery approach. METHODS AND RESULTS: MI was induced in mice and then randomized to targeted injection of an adenoviral construct (10 µL; 8×10(9) plaque forming units/mL) encoding green fluorescent protein (GFP) and the full-length human TIMP-4 (Ad-GFP-TIMP4) or GFP. A transgenic construct with cardiac-restricted overexpression TIMP-4 (hTIMP-4exp) was used in a parallel set of studies. LV end-diastolic volume, an index of LV remodeling, increased by >60% from baseline at 5 days post-MI and by >100% at 21 days post-MI in the Ad-GFP only group. However, LV dilation was reduced by ≈50% in both the Ad-GFP-TIMP4 and hTIMP-4exp groups at these post-MI time points. LV ejection fraction was improved with either Ad-GFP-TIMP-4 or hTIMP-4exp. Fibrillar collagen expression and content were increased within the MI region with both TIMP-4 interventions, suggestive of matrix stabilization. CONCLUSIONS: This study is the first to demonstrate that selective myocardial targeting for TIMP-4 induction through either a viral or transgenic approach favorably altered the course of adverse LV remodeling post-MI. Thus, localized induction of endogenous matrix metalloproteinase inhibitors, such as TIMP-4, holds promise as a means to interrupt the progression of post-MI remodeling.


Subject(s)
Gene Targeting , Gene Transfer Techniques , Myocardial Infarction/therapy , Tissue Inhibitor of Metalloproteinases/metabolism , Ventricular Function, Left , Ventricular Remodeling , Adenoviridae/genetics , Animals , Apoptosis , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Fibrillar Collagens/genetics , Fibrillar Collagens/metabolism , Gene Expression Regulation , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Transgenic , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , RNA, Messenger/metabolism , Recombinant Fusion Proteins/metabolism , Recovery of Function , Stroke Volume , Time Factors , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Inhibitor of Metalloproteinase-4
13.
J Biol Chem ; 286(19): 16976-83, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21367866

ABSTRACT

The microtubule-associated protein tau, which becomes hyperphosphorylated and pathologically aggregates in a number of these diseases, is extremely sensitive to manipulations of chaperone signaling. For example, Hsp90 inhibitors can reduce the levels of tau in transgenic mouse models of tauopathy. Because of this, we hypothesized that a number of Hsp90 accessory proteins, termed co-chaperones, could also affect tau stability. Perhaps by identifying these co-chaperones, new therapeutics could be designed to specifically target these proteins and facilitate tau clearance. Here, we report that the co-chaperone Cdc37 can regulate aspects of tau pathogenesis. We found that suppression of Cdc37 destabilized tau, leading to its clearance, whereas Cdc37 overexpression preserved tau. Cdc37 was found to co-localize with tau in neuronal cells and to physically interact with tau from human brain. Moreover, Cdc37 levels significantly increased with age. Cdc37 knockdown altered the phosphorylation profile of tau, an effect that was due in part to reduced tau kinase stability, specifically Cdk5 and Akt. Conversely, GSK3ß and Mark2 were unaffected by Cdc37 modulation. Cdc37 overexpression prevented whereas Cdc37 suppression potentiated tau clearance following Hsp90 inhibition. Thus, Cdc37 can regulate tau in two ways: by directly stabilizing it via Hsp90 and by regulating the stability of distinct tau kinases. We propose that changes in the neuronal levels or activity of Cdc37 could dramatically alter the kinome, leading to profound changes in the tau phosphorylation signature, altering its proteotoxicity and stability.


Subject(s)
Cell Cycle Proteins/chemistry , Chaperonins/chemistry , HSP90 Heat-Shock Proteins/metabolism , tau Proteins/chemistry , Alzheimer Disease/metabolism , Brain/metabolism , Cell Line, Tumor , HeLa Cells , Humans , Immunohistochemistry/methods , Molecular Chaperones/chemistry , Neurons/metabolism , Phosphorylation , RNA, Small Interfering/metabolism , Transfection
14.
Curr Neuropharmacol ; 9(4): 623-31, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22654720

ABSTRACT

The heat shock protein (Hsp) family is an evolutionarily conserved system that is charged with preventing unfolded or misfolded proteins in the cell from aggregating. In Alzheimer's disease, extracellular accumulation of the amyloid ß peptide (Aß) and intracellular aggregation of the microtubule associated protein tau may result from mechanisms involving chaperone proteins like the Hsps. Due to the ability of Hsps to regulate aberrantly accumulating proteins like Aß and tau, therapeutic strategies are emerging that target this family of chaperones to modulate their pathobiology. This article focuses on the use of Hsp-based therapeutics for treating primary and secondary tauopathies like Alzheimer's disease. It will particularly focus on the pharmacological targeting of the Hsp70/90 system and the value of manipulating Hsp27 for treating Alzheimer's disease.

15.
J Nat Prod ; 74(1): 38-44, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21141876

ABSTRACT

Target-based drug discovery for Alzheimer's disease (AD) centered on modulation of the amyloid ß peptide has met with limited success. Therefore, recent efforts have focused on targeting the microtubule-associated protein tau. Tau pathologically accumulates in more than 15 neurodegenerative diseases and is most closely linked with postsymptomatic progression in AD. We endeavored to identify compounds that decrease tau stability rather than prevent its aggregation. An extract from Myrica cerifera (bayberry/southern wax myrtle) potently reduced both endogenous and overexpressed tau protein levels in cells and murine brain slices. The bayberry flavonoids myricetin and myricitrin were confirmed to contribute to this potency, but a diarylheptanoid, myricanol, was the most effective anti-tau component in the extract, with potency approaching the best targeted lead therapies. (+)-aR,11S-Myricanol, isolated from M. cerifera and reported here for the first time as the naturally occurring aglycone, was significantly more potent than commercially available (±)-myricanol. Myricanol may represent a novel scaffold for drug development efforts targeting tau turnover in AD.


Subject(s)
Alzheimer Disease/drug therapy , Diarylheptanoids/isolation & purification , Diarylheptanoids/pharmacology , Flavonoids/isolation & purification , Flavonoids/pharmacology , Myrica/chemistry , tau Proteins/metabolism , Animals , Diarylheptanoids/chemistry , Female , Flavonoids/chemistry , HeLa Cells , Humans , Male , Mice , Models, Biological , Plant Roots/chemistry , Prosencephalon/cytology , Prosencephalon/drug effects , tau Proteins/analysis , tau Proteins/drug effects
16.
J Neurosci ; 30(46): 15374-82, 2010 Nov 17.
Article in English | MEDLINE | ID: mdl-21084594

ABSTRACT

Molecular chaperones regulate the aggregation of a number of proteins that pathologically misfold and accumulate in neurodegenerative diseases. Identifying ways to manipulate these proteins in disease models is an area of intense investigation; however, the translation of these results to the mammalian brain has progressed more slowly. In this study, we investigated the ability of one of these chaperones, heat shock protein 27 (Hsp27), to modulate tau dynamics. Recombinant wild-type Hsp27 and a genetically altered version of Hsp27 that is perpetually pseudo-phosphorylated (3×S/D) were generated. Both Hsp27 variants interacted with tau, and atomic force microscopy and dynamic light scattering showed that both variants also prevented tau filament formation. However, extrinsic genetic delivery of these two Hsp27 variants to tau transgenic mice using adeno-associated viral particles showed that wild-type Hsp27 reduced neuronal tau levels, whereas 3×S/D Hsp27 was associated with increased tau levels. Moreover, rapid decay in hippocampal long-term potentiation (LTP) intrinsic to this tau transgenic model was rescued by wild-type Hsp27 overexpression but not by 3×S/D Hsp27. Because the 3×S/D Hsp27 mutant cannot cycle between phosphorylated and dephosphorylated states, we can conclude that Hsp27 must be functionally dynamic to facilitate tau clearance from the brain and rescue LTP; however, when this property is compromised, Hsp27 may actually facilitate accumulation of soluble tau intermediates.


Subject(s)
HSP27 Heat-Shock Proteins/physiology , Molecular Dynamics Simulation , Neuronal Plasticity/genetics , tau Proteins/genetics , tau Proteins/metabolism , Animals , Female , Hippocampus/metabolism , Hippocampus/pathology , Male , Mice , Mice, Transgenic , Phosphorylation/physiology
17.
Mol Neurodegener ; 5: 45, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-21040568

ABSTRACT

BACKGROUND: It has traditionally been thought that the pathological accumulation of tau in Alzheimer's disease and other tauopathies facilitates neurodegeneration, which in turn leads to cognitive impairment. However, recent evidence suggests that tau tangles are not the entity responsible for memory loss, rather it is an intermediate tau species that disrupts neuronal function. Thus, efforts to discover therapeutics for tauopathies emphasize soluble tau reductions as well as neuroprotection. RESULTS: Here, we found that neuroprotection alone caused by methylene blue (MB), the parent compound of the anti-tau phenothiaziazine drug, Rember™, was insufficient to rescue cognition in a mouse model of the human tauopathy, progressive supranuclear palsy (PSP) and fronto-temporal dementia with parkinsonism linked to chromosome 17 (FTDP17): Only when levels of soluble tau protein were concomitantly reduced by a very high concentration of MB, was cognitive improvement observed. Thus, neurodegeneration can be decoupled from tau accumulation, but phenotypic improvement is only possible when soluble tau levels are also reduced. CONCLUSIONS: Neuroprotection alone is not sufficient to rescue tau-induced memory loss in a transgenic mouse model. Development of neuroprotective agents is an area of intense investigation in the tauopathy drug discovery field. This may ultimately be an unsuccessful approach if soluble toxic tau intermediates are not also reduced. Thus, MB and related compounds, despite their pleiotropic nature, may be the proverbial "magic bullet" because they not only are neuroprotective, but are also able to facilitate soluble tau clearance. Moreover, this shows that neuroprotection is possible without reducing tau levels. This indicates that there is a definitive molecular link between tau and cell death cascades that can be disrupted.

18.
Mol Cell Pharmacol ; 2(2): 43-46, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20523917

ABSTRACT

Neurodegenerative diseases caused by abnormal accumulation of the microtubule associated protein tau (MAPT, tau) are collectively called tauopathies. The most devastating tau related disorder is Alzheimer's disease (AD). Molecular chaperones such as heat shock proteins (Hsp) have emerged as critical regulators of tau stability. Several studies from our group and others have shown that the chaperone network can be targeted for the development of therapeutic strategies for AD and other neurodegenerative diseases. Here we will discuss a recent paper and current work from our laboratory where we have manipulated the ATPase activity of the 70-kDa heat shock protein (Hsp70) to regulate tau turnover. A high-throughput screening assay revealed several compounds that activated or inhibited Hsp70's ATPase activity. Inhibitors dramatically and rapidly reduced tau levels, whereas activators stabilized tau, both in cells and brain tissue. Moreover, increased levels of Hsp70 improved ATPase inhibitor efficacy, suggesting that therapies aimed at inducing Hsp70 levels followed by inhibition of its ATPase activity may be a very effective strategy to treat AD. These findings demonstrate that Hsp70 ATPase activity can be targeted to modify the pathologies of AD and other tauopathies.

19.
J Biol Chem ; 285(22): 16798-805, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20308058

ABSTRACT

The microtubule-associated protein Tau plays a crucial role in regulating the dynamic stability of microtubules during neuronal development and synaptic transmission. In a group of neurodegenerative diseases, such as Alzheimer disease and other tauopathies, conformational changes in Tau are associated with the initial stages of disease pathology. Folding of Tau into the MC1 conformation, where the amino acids at residues 7-9 interact with residues 312-342, is one of the earliest pathological alterations of Tau in Alzheimer disease. The mechanism of this conformational change in Tau and the subsequent effect on function and association to microtubules is largely unknown. Recent work by our group and others suggests that members of the Hsp70 family play a significant role in Tau regulation. Our new findings suggest that heat shock cognate (Hsc) 70 facilitates Tau-mediated microtubule polymerization. The association of Hsc70 with Tau was rapidly enhanced following treatment with microtubule-destabilizing agents. The fate of Tau released from the microtubule was found to be dependent on ATPase activity of Hsc70. Microtubule destabilization also rapidly increased the MC1 folded conformation of Tau. An in vitro assay suggests that Hsc70 facilitates formation of MC1 Tau. However, in a hyperphosphorylating environment, the formation of MC1 was abrogated, but Hsc70 binding to Tau was enhanced. Thus, under normal circumstances, MC1 formation may be a protective conformation facilitated by Hsc70. However, in a diseased environment, Hsc70 may preserve Tau in a more unstructured state, perhaps facilitating its pathogenicity.


Subject(s)
HSC70 Heat-Shock Proteins/metabolism , Microtubules/metabolism , tau Proteins/chemistry , Animals , Chaperonins/chemistry , HeLa Cells , Humans , Immunohistochemistry/methods , Microscopy, Fluorescence/methods , Microtubules/chemistry , Models, Biological , Oocytes/metabolism , Phosphorylation , Protein Binding , Recombinant Proteins/chemistry , Xenopus
20.
J Neurosci ; 30(2): 591-9, 2010 Jan 13.
Article in English | MEDLINE | ID: mdl-20071522

ABSTRACT

Imbalanced protein load within cells is a critical aspect for most diseases of aging. In particular, the accumulation of proteins into neurotoxic aggregates is a common thread for a host of neurodegenerative diseases. Our previous work demonstrated that age-related changes to the cellular chaperone repertoire contributes to abnormal buildup of the microtubule-associated protein tau that accumulates in a group of diseases termed tauopathies, the most common being Alzheimer's disease. Here, we show that the Hsp90 cochaperone, FK506-binding protein 51 (FKBP51), which possesses both an Hsp90-interacting tetratricopeptide domain and a peptidyl-prolyl cis-trans isomerase (PPIase) domain, prevents tau clearance and regulates its phosphorylation status. Regulation of the latter is dependent on the PPIase activity of FKBP51. FKB51 enhances the association of tau with Hsp90, but the FKBP51/tau interaction is not dependent on Hsp90. In vitro FKBP51 stabilizes microtubules with tau in a reaction depending on the PPIase activity of FKBP51. Based on these new findings, we propose that FKBP51 can use the Hsp90 complex to isomerize tau, altering its phosphorylation pattern and stabilizing microtubules.


Subject(s)
Brain/metabolism , HSP90 Heat-Shock Proteins/metabolism , Microtubules/metabolism , Tacrolimus Binding Proteins/metabolism , tau Proteins/metabolism , Animals , Benzoquinones/pharmacology , Cell Line, Transformed , Chymotrypsin/pharmacology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Lactams, Macrocyclic/pharmacology , Male , Mice , Mice, Inbred C57BL , Microtubules/drug effects , Mutation/genetics , Oocytes , Peptidylprolyl Isomerase/metabolism , RNA, Small Interfering/pharmacology , Tacrolimus Binding Proteins/genetics , Transfection/methods , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...