Wednesday, January 4, 2012

tau tangles| What is tau tangles|Papers on tau tangles |Research on tau tangles | Publications on tau tangles


1.
Neural Plast. 2012;2012:319836. Epub 2011 Nov 28.

Spines, plasticity, and cognition in Alzheimer's model mice.

Source

Massachusetts General Hospital, Harvard Medical School, 114 16th Street, Charlestown, MA 02129, USA.

Abstract

The pathological hallmarks of Alzheimer's disease (AD)-widespread synaptic and neuronal loss and the pathological accumulation of amyloid-beta peptide (Aβ) in senile plaques, as well as hyperphosphorylated tau in neurofibrillarytangles-have been known for many decades, but the links between AD pathology and dementia and effective therapeutic strategies remain elusive. Transgenic mice have been developed based on rare familial forms of AD and frontotemporal dementia, allowing investigators to test in detail the structural, functional, and behavioral consequences of AD-associated pathology. Here, we review work on transgenic AD models that investigate the degeneration of dendritic spine structure, synaptic function, and cognition. Together, these data support a model of AD pathogenesis in which soluble Aβ initiates synaptic dysfunction and loss, as well as pathological changes in tau, which contribute to both synaptic and neuronal loss. These changes in synapse structure and function as well as frank synapse and neuronal loss contribute to the neural system dysfunction which causes cognitive deficits. Understanding the underpinnings of dementia in AD will be essential to develop and evaluate therapeutic approaches for this widespread and devastating disease.

PMID:
22203915
[PubMed - in process]
PMCID: PMC3238410
Free PMC Article
Click here to read
2.
Front Biosci (Elite Ed). 2012 Jan 1;4:2101-9.

The contributions of unscheduled neuronal cell cycle events to the death of neurons in Alzheimer's disease.

Source

Dept. of Cell Biology and Neuroscience, Nelson Biological Laboratories, 604 Allison Road, Piscataway, NJ 0854.

Abstract

Alzheimer's disease is a neurodegenerative disorder that accounts for the majority of the dementia in individuals over the age of 65. While much has been learned about the biology and biochemistry of the tau tangles and beta-amyloid plaques, less is known about the cell biology of the neuronal cell death process. This review examines one feature of this process, the unexpected occurrence of unscheduled cell cycle events in mature and normally non-mitotic neurons in the at-risk neuronal populations. The correlation of neuronal cell cycling and cell death is not unique to Alzheimer's, but the evidence in both human Alzheimer's disease and its mouse models suggests that these events are early disease related processes, that they are driving forces of the disease rather than indirect symptoms. Defining the biochemistry behind cell cycle initiation holds promise as a fresh therapeutic approach in the battle against this devastating disease.

PMID:
22202022
[PubMed - in process]
3.
Pharmacol Ther. 2011 Dec 16. [Epub ahead of print]

Pharmacotherapies for Alzheimer's disease: Beyond cholinesterase inhibitors.

Source

Department of Neurology, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Belmont, MA 02478, USA; Department of Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.

Abstract

Alzheimer's disease (AD) is the most common cause of memory impairment and dementia in the elderly. AD is pathologically characterized by extracellular deposits of beta-amyloid (Aβ) peptide, neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau, neuronal loss, and neurotransmitter dysfunction. Clinically, AD is characterized by progressive cognitive decline that usually starts with memory impairment and progresses to cause a more generalized cognitive dysfunction, behavioral dysregulation, and neuropsychiatric symptoms. These symptoms collectively lead to a progressive and relentless decline in the ability to perform functions of daily living, eventually leading to total incapacitation. The incidence and prevalence of AD are expected to exponentially increase with the aging of the population. Currently approved treatments, including the acetylcholinesterase inhibitors (AChEIs) donepezil, galantamine and rivastigmine, and the N-methyl-D-aspartate (NMDA) antagonist memantine, do not halt the progression of the disease, and have provided marginal therapeutic benefits. Accordingly, there is an urgent need to develop novel and effective medications for AD that go beyond AChEIs and NMDA antagonists. Modern research has focused on discovering effective disease-modifying therapies, which specifically target the pathophysiologic cascade, hoping to delay the onset of the disease and slow its progression. In this review, different pharmacological drugs and therapeutic approaches will be discussed, with an emphasis on novel therapies that are currently being investigated in clinical trials.

Copyright © 2011. Published by Elsevier Inc.

PMID:
22198801
[PubMed - as supplied by publisher]
Click here to read
4.
Neurology. 2011 Dec 21. [Epub ahead of print]

Amyloid pathology in persons with "normal" cognition.

Source

From the Rush Alzheimer's Disease Center, Department of Neurological Sciences, Rush University Medical Center, Chicago, IL.

Abstract

Alzheimer disease (AD) is characterized pathologically by cortical neuritic plaques composed of a central core of amyloid-β (Aβ) peptide fibrils and neurofibrillary tangles composed of abnormally phosphorylated tau. The deposition of Aβ is thought to be an early step in a cascade of events that ultimately leads to the loss of cognitive abilities and dementia that characterizes AD. Thus, it was not surprising to find AD pathology in persons with mild cognitive impairment (MCI).(1) Clinical-pathologic studies have also reported a relation between AD pathology and cognition in persons without MCI or dementia.(2) Over the past several years, clinical-pathologic studies have been confirmed with in vivo imaging of amyloid by PET using Pittsburgh compound B (PiB).(3) Overall, these findings have led to a new conceptualization of AD as beginning with an asymptomatic pathophysiologic process, followed by a MCI stage, and ultimately the final AD dementia stage.(4,5) Understanding factors that influence the onset and progression of AD is of great interest. The apolipoprotein E ε4 allele polymorphism (APOE) is the most important genetic risk factor for AD and may increase disease risk, in part, by influencing Aβ clearance and fibrillogenesis.(6) In fact, some clinical-pathologic data suggest that Aβ mediates the association of APOE with cognition, at least in studies that include the full range of cognition.(7) However, the influence of APOE on Aβ and cognition among persons without cognitive impairment has not been well studied. In this issue of Neurology®, Kantarci et al.(8) use data from the Mayo Clinic Study of Aging (MCSA) to examine the relation of APOE to Aβ identified by PiB PET imaging and cognitive function in persons without cognitive impairment.

PMID:
22189449
[PubMed - as supplied by publisher]
Click here to read
5.
J Biol Chem. 2011 Dec 19. [Epub ahead of print]

Propyl isomerase Pin1 promotes APP protein turnover by inhibiting GSK3β kinase activity: A novel mechanism for Pin1 to protect against Alzheimer's disease.

Source

Beth Israel Deaconese Medical Center, United States;

Abstract

Alzheimer's disease (AD) is characterized by the presence of senile plaques of amyloid-beta peptides (Aβ) derived from amyloid precursor protein (APP) and neurofibrillary tangles composed of hyperphosphorylated tau. Increasing APP gene dosage or expression has been shown to cause familial early-onset AD. However, whether protein stability of APP is regulated is unclear. The prolyl isomerase Pin1 and glycogen synthase 3β (GSK3β) have been shown to have the opposite effects on APP processing and tau hyperphosphorylation, relevant to the pathogenesis of AD. However, nothing is known about their relationship. In this study, we found that Pin1 binds to the pT330-P motif in GSK3β to inhibit its kinase activity. Furthermore, Pin1 promotes protein turnover of APP by inhibiting GSK3β activity. A point mutation either at T330, the Pin1-binding site in GSK3β, or T668, the GSK3β phosphorylation site in APP, abolished the regulation of GSK3β activity, T668 phosphorylation and APP stability by Pin1, resulting in reduced non-amyloidogenic APP processing and increased APP levels. These results uncover a novel role of Pin1 in inhibiting GSK3β kinase activity to reduce APP protein levels, providing a previously unrecognized mechanism by which Pin1 protects against Alzheimer's disease.

PMID:
22184106
[PubMed - as supplied by publisher]
Free full text
Click here to read
6.
Neurocase. 2011 Dec 20. [Epub ahead of print]

Autopsy-proven progressive supranuclear palsy presenting as behavioral variant frontotemporal dementia.

Source

a Department of Neurology , Mayo Clinic , Rochester , MN , USA.

Abstract

Background: Progressive supranuclear palsy (PSP) is a neurodegenerative disorder pathologically characterized by neuronal loss, gliosis and tau-positive neurofibrillary tangles in basal ganglia, brainstem and cerebellar nuclei. Five presenting clinical syndromes of PSP are well-described: (i) the classic Richardson's syndrome; (ii) asymmetric parkinsonism with tremor; (iii) freezing of gait; (iv) asymmetric limb apraxia, and (v) apraxia of speech. Aim: To determine whether autopsy-proven PSP cases may present with another clinical phenotype. Methods: Medical records of 66 autopsy-proven PSP cases between 1973 and 2010 were reviewed to determine whether all could be classified into one of five well-defined syndromes listed above. Three cases presented with prominent behavioral and personality changes, meeting diagnosis of behavioral variant frontotemporal dementia. MRI midbrain and pons volumes and pons/midbrain ratios were compared to healthy controls and typical PSP cases. Results: All three bvFTD cases developed at least one PSP symptom or sign that emerged up to 5 years after initial presentation. One case was re-diagnosed as PSP 6 years after presentation as bvFTD. Compared to controls, midbrain volume was significantly smaller in both bvFTD (p = .03) and PSP cases (p = .008), without significant difference between PSP and bvFTD cases (.44). However pontine volumes were similar across all three groups. Conclusions: While most autopsy-confirmed PSP cases present with one of the five well-described syndromes, there are cases that may present as bvFTD. In these, at least one cardinal symptom or sign of PSP later emerges, associated with smaller midbrain volume and increased pons/midbrain ratio. Thus underlying PSP pathology should be considered in these cases.

PMID:
22181323
[PubMed - as supplied by publisher]
Click here to read
7.
PLoS One. 2011;6(12):e26860. Epub 2011 Dec 8.

Tau-Targeted Immunization Impedes Progression of Neurofibrillary Histopathology in Aged P301L Tau Transgenic Mice.

Source

Laboratory for Translational Neurodegeneration, Brain and Mind Research Institute, The University of Sydney, Camperdown, New South Wales, Australia.

Abstract

In Alzheimer's disease (AD) brains, the microtubule-associated protein tau and amyloid-β (Aβ) deposit as intracellular neurofibrillary tangles (NFTs) and extracellular plaques, respectively. Tau deposits are furthermore found in a significant number of frontotemporal dementia cases. These diseases are characterized by progressive neurodegeneration, the loss of intellectual capabilities and behavioral changes. Unfortunately, the currently available therapies are limited to symptomatic relief. While active immunization against Aβ has shown efficacy in both various AD mouse models and patients with AD, immunization against pathogenic tau has only recently been shown to prevent pathology in young tautransgenic mice. However, if translated to humans, diagnosis and treatment would be routinely done when symptoms are overt, meaning that the histopathological changes have already progressed. Therefore, we used active immunization to target pathogenic tau in 4, 8, and 18 months-old P301L tau transgenic pR5 mice that have an onset of NFT pathology at 6 months of age. In all age groups, NFT pathology was significantly reduced in treated compared to control pR5 mice. Similarly, phosphorylation of tau at pathological sites was reduced. In addition, increased astrocytosis was found in the oldest treated group. Taken together, our data suggests that tau-targeted immunization slows the progression of NFT pathology in mice, with practical implications for human patients.

PMID:
22174735
[PubMed - in process]
PMCID: PMC3234245
Free PMC Article
Click here to readClick here to read
8.
Neurodegener Dis. 2011 Dec 7. [Epub ahead of print]

Linking Amyloid-β and Tau: Amyloid-β Induced Synaptic Dysfunction via Local Wreckage of the Neuronal Cytoskeleton.

Source

Max Planck Unit for Structural Molecular Biology, Hamburg, Germany.

Abstract

Background: In Alzheimer's disease (AD), amyloid-β (Aβ) is the major component of extracellular plaques, whereas the microtubule-associated protein tau forms the main component of intracellular tangles. In contrast to frontotemporal dementias and other neurodegenerative diseases, both proteins form pathological aggregates and are considered key players for the development of AD. However, the connection between Aβ and tau and the functional loss of neurons and synapses, which ultimately lead to cognitive impairments, is still not well understood. Objectives: Making use of primary neurons exposed to Aβ oligomers, we sought to determine how tau mediates the Aβ-induced neuronal dysfunction. Additionally, we asked how the microtubule cytoskeleton is involved in the combined Aβ and tau toxicity. Methods: We exposed mature primary rat neurons with developed synapses to Aβ oligomers and used immunofluorescence and electron microscopy to investigate tau, actin, neurofilament and microtuble cytoskeleton changes. Results: Aβ oligomers preferentially associate with synapses, notably dendritic spines, throughout the neuronal cell culture. As a consequence, endogenous tau gets missorted from the axonal into the somatodendritic compartment in a subset of cells. These missorted cells also display missorting of neurofilaments, and a dramatic loss of microtubules, which can be prevented by the microtubule stabilizer taxol. Conclusions: Aβ causes tau missorting, loss of neuronal cell polarity and loss of dendritic microtubules. This in turn leads to impaired organelle/mitochondria transport, whereby synapses cannot be maintained properly and eventually decay. The data support the view that the microtubule cytoskeleton is a valid therapeutic target in AD.

Copyright © 2011 S. Karger AG, Basel.

PMID:
22156588
[PubMed - as supplied by publisher]
Click here to read
9.
Neurosci Lett. 2012 Jan 17;507(1):22-6. Epub 2011 Dec 2.

Enriched odor exposure decrease tau phosphorylation in the rat hippocampus and cortex.

Source

Department of Pathophysiology, Key Laboratory of Neurological Diseases of Education Committee of China, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.

Abstract

Abnormally hyperphosphorylated microtubule-associated protein tau is the main component of the neurofibrillary tangles(NFT), a hallmark pathological feature of Alzheimer's disease (AD). A lot of studies suggested that there is highly neurobiological correlation between olfactory dysfunction and AD-like pathology, but the effect of the odor stimulation ontau phosphorylation remains unknown. Here, we examined the effect of short-term and long-term enriched odor exposure on the alterations of tau phosphorylation at multiple sites in the rat brains. We found that short-term odor enrichment did not affect the phosphorylation of tau, while long-term odor enrichment dramatically reduce the phosphorylation level oftau at Ser198/199/202, Thr231, Ser396, and Ser404 sites both in the hippocampus and cortex. These data suggest that long-term odor exposure prevent tau phosphorylation and may be a new therapeutic strategy of AD.

Copyright © 2011 Elsevier Ireland Ltd. All rights reserved.

PMID:
22155098
[PubMed - in process]
Click here to read
10.
Am J Pathol. 2011 Dec 3. [Epub ahead of print]

Rac1b Increases with Progressive Tau Pathology within Cholinergic Nucleus Basalis Neurons in Alzheimer's Disease.

Source

Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois.

Abstract

Cholinergic basal forebrain (CBF) nucleus basalis (NB) neurons display neurofibrillary tangles (NFTs) during Alzheimer's disease (AD) progression, yet the mechanisms underlying this selective vulnerability are currently unclear. Rac1, a member of the Rho family of GTPases, may interact with the proapoptotic pan-neurotrophin receptor p75(NTR) to induce neuronal cytoskeletal abnormalities in AD NB neurons. Herein, we examined the expression of Rac1b, a constitutively active splice variant of Rac1, in NB cholinergic neurons during AD progression. CBF tissues harvested from people who died with a clinical diagnosis of no cognitive impairment (NCI), mild cognitive impairment, or AD were immunolabeled for both p75(NTR) and Rac1b. Rac1b appeared as cytoplasmic diffuse granules, loosely aggregated filaments, or compact spheres in p75(NTR)-positive NB neurons. Although Rac1b colocalized with tau cytoskeletal markers, the percentage of p75(NTR)-immunoreactive neurons expressing Rac1b was significantly increased only in AD compared with both mild cognitive impairment and NCI. Furthermore, single-cell gene expression profiling with custom-designed microarrays showed down-regulation of caveolin 2, GNB4, and lipase A in AD Rac1b-positive/p75(NTR)-labeled NB neurons compared with Rac1b-negative/p75(NTR)-positive perikarya in NCI. These proteins are involved in Rac1 pathway/cell cycle progression and lipid metabolism. These data suggest that Rac1b expression acts as a modulator or transducer of various signaling pathways that lead to NFT formation and membrane dysfunction in a subgroup of CBF NB neurons in AD.

Copyright © 2012 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.

PMID:
22142809
[PubMed - as supplied by publisher]
Click here to read
11.
Prog Neurobiol. 2011 Nov 22;96(1):87-95. [Epub ahead of print]

Emerging role of p62/sequestosome-1 in the pathogenesis of Alzheimer's disease.

Source

Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland; Department of Neurology, Kuopio University Hospital, P.O. Box 1777, FIN-70211 Kuopio, Finland.

Abstract

The p62/sequestosome-1 is a multifunctional protein containing several protein-protein interaction domains. Through these interactions p62 is involved in the regulation of cellular signaling and protein trafficking, aggregation and degradation. p62 protein can bind through its UBA motif to ubiquitinated proteins and control their aggregation and degradation via either autophagy or proteasomes. p62 protein has been reported to be seen in association with the intracellular inclusions in primary and secondary tauopathies, α-synucleinopathies and other neurodegenerative brain disorders displaying inclusions with misfolded proteins. In Alzheimer's disease (AD), p62 protein is associated with neurofibrillary tangles composed primarily of hyperphosphorylated tau protein and ubiquitin. Increasing evidence indicates that p62 has an important role in the degradation of tau protein. The lack of p62 protein expression provokes the tau pathology in mice. Recent studies have demonstrated that the p62 gene expression and cytoplasmic p62 protein levels are significantly reduced in the frontal cortex of AD patients. Decline in the level of p62 protein can disturb the signaling pathways of Nrf2, cyclic AMP and NF-κB and in that way increase oxidative stress and impair neuronal survival. We will review here the molecular and functional characteristics of p62 protein and outline its potential role in the regulation of Alzheimer's pathogenesis.

Copyright © 2011 Elsevier Ltd. All rights reserved.

PMID:
22138392
[PubMed - as supplied by publisher]
Click here to read
12.
Alzheimers Dement. 2011 Nov 30. [Epub ahead of print]

Some evolutionary perspectives on Alzheimer's disease pathogenesis and pathology.

Source

Department of Psychology, State University of New York at New Paltz, New Paltz, NY, USA.

Abstract

There is increasing urgency to develop effective prevention and treatment for Alzheimer's disease (AD) as the aging population swells. Yet, our understanding remains limited for the elemental pathophysiological mechanisms of AD dementia that may be causal, compensatory, or epiphenomenal. To this end, we consider AD and why it exists from the perspectives of natural selection, adaptation, genetic drift, and other evolutionary forces. We discuss the connection between the apolipoprotein E (APOE) allele and AD, with special consideration to APOE ɛ4 as the ancestral allele. The phylogeny of AD-like changes across species is also examined, and pathology and treatment implications of AD are discussed from the perspective of evolutionary medicine. In particular, amyloid-β (Aβ) neuritic plaques and paired helical filament tau (PHFtau) neurofibrillary tangles have been traditionally viewed as injurious pathologies to be targeted, but may be preservative or restorative processes that mitigate harmful neurodegenerative processes or may be epiphenoma of the essential processes that cause neurodegeneration. Thus, we raise fundamental questions about current strategies for AD prevention and therapeutics.

Copyright © 2011 The Alzheimer's Association. Published by Elsevier Inc. All rights reserved.

PMID:
22137143
[PubMed - as supplied by publisher]
13.
Med Hypotheses. 2012 Feb;78(2):293-8. Epub 2011 Dec 2.

Dietary intake of cottonseed toxins is hypothesized to be a partial cause of Alzheimer's disorder.

Source

Wisconsin Institute of Nutrition, 6789 N. Green Bay Ave., Milwaukee, WI 53209, United States.

Abstract

The cause of Alzheimer's disorder is not known. The most influential known risk factor is increasing age. The risk factor of increasing age is consistent with exposure to environmental toxins throughout life as a cause of Alzheimer's. In addition, microbleeding, changes in membrane permeability and increased cholesterol are all factors important in Alzheimer's. Cottonseed contains toxins and is fed to animals, fish and poultry. Cottonseed toxins remain in the animals, fish and poultry and are present in the human diet at seemingly low levels. The average person is ingesting cottonseed toxins throughout life. Cottonseed toxins cause bleeding, changes in membrane permeability and increased cholesterol. In addition, the cottonseed toxin gossypol is known to reach the brain and bind randomly to important cellular structures. Gossypol also binds to microtubules and interferes with microtubule assembly, which may inhibit binding of tau to microtubules and lead to formation of neurofibrillary tangles. Cottonseed toxins are also known to accumulate in the body. In a preliminary study of female rats fed low level cottonseed for their lifetimes, apparent neurofibrillary tangles and phosphorylated tau were found. The intake of cottonseed toxins throughout life should be evaluated further as a possible cause of Alzheimer's.

Copyright © 2011 Elsevier Ltd. All rights reserved.

PMID:
22136946
[PubMed - in process]
Click here to read
14.
Antioxid Redox Signal. 2011 Nov 28. [Epub ahead of print]

Alzheimer's Disease: Redox Dysregulation as a Common Denominator for Diverse Pathogenic Mechanisms.

Source

Pontificia Universidad Catolica de Chile School of Medicine, Neurology, Marcoleta 391, Santiago, Chile, 8330024, 56-2-3546936, 56-2-6321924; rvonb@med.puc.cl.

Abstract

Significance: Alzheimer's disease (AD) is the most common cause of dementia and a progressive neurodegeneration that appears to result from multiple pathogenic mechanisms, including protein misfolding/ aggregation, involved in both amyloid β (Aβ)-dependent senile plaques and tau-dependent neurofibrillary tangles; metabolic and mitochondrial dysfunction, excitoxicity, calcium handling impairment, glial cell dysfunction, neuroinflammation and oxidative stress. Oxidative stress, which could be secondary to several of the other pathophysiological mechanisms, appears as a major determinant of the pathogenesis and progression of AD. Recent advances: The identification of oxidized proteins common for mild cognitive impairment and AD suggests that key oxidation pathways are triggered early and are involved in the initial progression of the neurodegenerative process. Abundant data support that oxidative stress, also considered as a main factor for aging, the major risk factor for AD, can be a common key element capable of articulating the divergent nature of the proposed pathogenic factors. Critical issues: Pathogenic mechanisms influence each other at different levels. Evidence suggests that it will be difficult to define a single target therapy resulting in the arrest of progression or the improvement of AD deterioration. Since oxidative stress is present from early stages of disease, it appears as one of the main targets to be included in a clinical trial. Future Directions: Exploring the articulation of AD pathogenic mechanisms by oxidative stress will provide clues for better understanding the pathogenesis and progression of this dementing disorder and for the development of effective therapies to treat this disease.

PMID:
22122400
[PubMed - as supplied by publisher]
Click here to read
15.
Antioxid Redox Signal. 2011 Nov 27. [Epub ahead of print]

Insights into Mitochondrial Dysfunction: Aging, Amyloid-β and Tau - a deleterious trio.

Source

Universitäre Psychiatrische Kliniken Basel, Neurobiology Lab for Brain Aging & Mental Health, Basel, Switzerland; karen.schmitt@upkbs.ch.

Abstract

Significance. Alzheimer's disease (AD) is an age-related progressive neurodegenerative disorder mainly affecting elderly individuals. The pathology of AD is characterized by amyloid plaques (aggregates of amyloid-β (Aβ)) and neurofibrillarytangles (aggregates of tau), but the mechanisms underlying this dysfunction are still partially unclear. Recent Advances. A growing body of evidence supports mitochondrial dysfunction as a prominent and early, chronic oxidative stress-associated event that contributes to synaptic abnormalities and, ultimately, selective neuronal degeneration in AD. Critical issues. In this review, we discuss on the one hand whether mitochondrial decline observed in brain aging is a determinant event in the onset of AD and on the other hand the close inter-relationship of this organelle with Aβ and tauin the pathogenic process underlying AD. Moreover, we summarize evidence from aging and Alzheimer models showing that the harmful trio "aging, Aβ and tau protein" triggers mitochondrial dysfunction through a number of pathways, such as impairment of oxidative phosphorylation, elevation of reactive oxygen species production and interaction with mitochondrial proteins, contributing to the development and progression of the disease. Future Directions. The aging process may weaken the mitochondrial OXPHOS system in a more general way over many years providing a basis for the specific and destructive effects of Aβ and tau. Establishing strategies involving efforts to protect cells at the mitochondrial level by stabilizing or restoring mitochondrial function and energy homeostasis appears to be challenging, but very promising route on the horizon.

PMID:
22117646
[PubMed - as supplied by publisher]
Click here to read
16.
Histochem Cell Biol. 2011 Nov 25. [Epub ahead of print]

Tangle evolution linked to differential 3- and 4-repeat tau isoform deposition: a double immunofluorolabeling study using two monoclonal antibodies.

Source

Laboratory of Structural Neuropathology, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya, Tokyo, 156-8509, Japan, uchihara-ts@igakuken.or.jp.

Abstract

Double immunofluorolabeling for 3-repeat (3R) and 4-repeat (4R) tau was performed with two monoclonal antibodies, RD3 and RD4, after an additional pretreatment with potassium permanganate and oxalic acid to eliminate nonspecific 3R taucytoplasmic staining. This method involves hyperdilution of one of the primary monoclonal antibodies (≥100-fold), making it undetectable by usual secondary antibodies. The hyperdiluted primary antibody can then only be detected after tyramide amplification. Subsequent application of the other monoclonal antibody at its usual concentration allows double immunofluorolabeling without cross-reaction. This novel method revealed that tau immunoreactivity (IR) in the hippocampal pyramidal neurons of Alzheimer's disease (AD) brains is heterogeneous in that pretangle neurons exhibit 4R-selective (3R-/4R+) IR, ghost tangles exhibit 3R-selective (3R+/4R-) IR, and neurofibrillary tangles exhibit both 3R and 4R (3R+/4R+) IR. Some nigral neurons exhibited RD3 IR in both AD and corticobasal degeneration/progressive supranuclear palsy (CBD/PSP) brains. However, in CBD/PSP cases, 3R IR was always superimposed on 4R IR, while 3R-selective neurons were present in AD cases. These differential isoform profiles may provide a pivotal molecular reference, closely related to the morphological evolution of tau-positive neurons, which may be variable according to disease (CBD/PSP vs. AD), lesion site (cerebral cortex and substantia nigra), or the stage of evolution (from pretangles to ghost tangles). These findings should provide a more comprehensive understanding of the histological differentiation of various tau deposits in human neurodegenerative disease.

PMID:
22116524
[PubMed - as supplied by publisher]
Click here to read
17.
Exp Neurobiol. 2011 Jun;20(2):67-80. Epub 2011 Jun 30.

The Interface between Cytoskeletal Aberrations and Mitochondrial Dysfunction in Alzheimer's Disease and Related Disorders.

Source

Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.

Abstract

The major defining pathological hallmarks of Alzheimer's disease (AD) are the accumulations of Aβ in senile plaques and hyperphosphorylated tau in neurofibrillary tangles and neuropil threads. Recent studies indicate that rather than these insoluble lesions, the soluble Aβ oligomers and hyperphosphorylated tau are the toxic agents of AD pathology. Such pathological protein species are accompanied by cytoskeletal changes, mitochondrial dysfunction, Ca(2+) dysregulation, and oxidative stress. In this review, we discuss how the binding of Aβ to various integrins, defects in downstream focal adhesion signaling, and activation of cofilin can impact mitochondrial dysfunction, cytoskeletal changes, and tau pathology induced by Aβ oligomers. Such pathological consequences can also feedback to further activate cofilin to promote cofilin pathology. We also suggest that the mechanism of Aβ generation by the endocytosis of APP is mechanistically linked with perturbations in integrin-based focal adhesion signaling, as APP, LRP, and β-integrins are physically associated with each other.

PMID:
22110363
[PubMed]
PMCID: PMC3213703
Free PMC Article
Click here to readClick here to read
18.
Exp Neurobiol. 2010 Dec;19(3):120-31. Epub 2010 Dec 31.

Drug development for Alzheimer's disease: recent progress.

Source

Institute for Brain Science and Technology (IBST)/Graduate Program in Neuroscience, Inje University, Busan 614-735, Korea.

Abstract

Alzheimer's disease, the most common cause of dementia, is characterized by two major pathological hallmarks: amyloid plaques and neurofibrillary tangles. Based on these two indicators, an amyloid cascade hypothesis was proposed, and accordingly, most current therapeutic approaches are now focused on the removal of β-amyloid peptides (Aβ from the brain. Additionally, strategies for blocking tau hyperphosphorylation and aggregation have been suggested, including the development of drugs that can block the formation of tangles. However, there are no true disease-modifying drugs in the current market, though many drugs based on theories other than Aβ and tau pathology are under development. The purpose of this review was to provide information on the current development of AD drugs and to discuss the issues related to drug development.

PMID:
22110351
[PubMed]
PMCID: PMC3214787
Free PMC Article
Click here to readClick here to read
19.
PLoS One. 2011;6(11):e27068. Epub 2011 Nov 11.

Abnormal cognition, sleep, EEG and brain metabolism in a novel knock-in Alzheimer mouse, PLB1.

Source

School of Medical Sciences, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen, United Kingdom. b.platt@abdn.ac.uk

Abstract

Late-stage neuropathological hallmarks of Alzheimer's disease (AD) are β-amyloid (βA) and hyperphosphorylated taupeptides, aggregated into plaques and tangles, respectively. Corresponding phenotypes have been mimicked in existing transgenic mice, however, the translational value of aggressive over-expression has recently been questioned. As controlled gene expression may offer animal models with better predictive validity, we set out to design a transgenic mouse model that circumvents complications arising from pronuclear injection and massive over-expression, by targeted insertion of human mutated amyloid and tau transgenes, under the forebrain- and neurone-specific CaMKIIα promoter, termed PLB1(Double). Crossing with an existing presenilin 1 line resulted in PLB1(Triple) mice. PLB1(Triple) mice presented with stable gene expression and age-related pathology of intra-neuronal amyloid and hyperphosphorylated tauin hippocampus and cortex from 6 months onwards. At this early stage, pre-clinical (18)FDG PET/CT imaging revealed cortical hypometabolism with increased metabolic activity in basal forebrain and ventral midbrain. Quantitative EEG analyses yielded heightened delta power during wakefulness and REM sleep, and time in wakefulness was already reliably enhanced at 6 months of age. These anomalies were paralleled by impairments in long-term and short-term hippocampal plasticity and preceded cognitive deficits in recognition memory, spatial learning, and sleep fragmentation all emerging at ∼12 months. These data suggest that prodromal AD phenotypes can be successfully modelled in transgenic mice devoid of fibrillary plaque or tangle development. PLB1(Triple) mice progress from a mild (MCI-like) state to a more comprehensive AD-relevant phenotype, which are accessible using translational tools such as wireless EEG and microPET/CT.

PMID:
22096518
[PubMed - in process]
PMCID: PMC3214038
Free PMC Article
Click here to readClick here to read
20.
J Neurogenet. 2011 Dec;25(4):127-33.

Targeting post-translational modifications on tau as a therapeutic strategy for Alzheimer's disease.

Source

Department of Neurosymptomatics, Merck Research Laboratories, West Point, PA 19486, USA. jacob_marcus@merck.com

Abstract

Alzheimer's disease (AD) is a neurodegenerative disorder that causes early memory impairment, followed by profound progressive cognitive decline, and eventually death. Neurofibrillary tangles (NFTs) are one of the histopathological hallmarks of AD. NFTs are deposits of insoluble aggregates of the microtubule-binding protein tau, left behind following neuronal loss. Intracellular aggregates of tau, either in soluble or insoluble forms, are thought to disrupt cellular machinery and synaptic function and ultimately lead to neuronal death. As the ultimate pathological endpoint in AD is neuronal loss, there is significant interest in understanding the causes of tau aggregation and deposition in the brain as a potential therapeutic avenue for AD. Post-translational modifications on tau are thought to be an important regulatory mechanism that may contribute to the propensity of tau to aggregate and form NFTs. In addition to phosphorylation, numerous other post-translational modifications have been observed on tau protein. The mechanisms that cause aggregation of tau are unknown, but it is likely that post-translational modifications other than phosphorylation also regulate this process. This review will discuss several post-translational modifications of tau and their roles in modulation of tau function and aggregation in AD.

No comments:

Post a Comment