Tuesday, September 6, 2011

Tau Tangles~ Current Research TOP 50 Publications on Tau Tangles~ Tau Proteins Blog

Tau Tangles~ Current Research TOP 50 Publications on Tau Tangles~ Tau Proteins Blog

Epilepsia. 2007 Aug;48(8):1447-54. Epub 2007 Apr 18.
Source
Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, Queen Square, London, UK.
Abstract
PURPOSE:
Reactivation of neurodevelopmental processes may contribute to neurodegeneration. For example, the proteins cyclin dependent kinase 5 (cdk5) and glycogen synthase kinase 3 beta (GSK3beta), which are essential to normal cortical development, can hyperphosphorylate tau and might contribute to the pathogenesis of Alzheimer's disease. Focal cortical dysplasia (FCD) is an important neurodevelopmental cause of refractory human epilepsy within which dysplastic neurons exhibit increased immunoreactivity for cdk5 and GSK3beta as well as neurofilamentous accumulations. We therefore hypothesized that the developmentally abnormal cortex of FCD might be more susceptible to tau-mediated neurodegeneration than adjacent histologically normal cortex.
MATERIALS AND METHODS:
We examined a series of 15 cases of FCD, spanning a wide age range, for beta-amyloid, pathologically phosphorylated tau and neurofibrillary tangles using silver staining, immunohistochemistry for tau, AT8, RD3, RD4 and two-dimensional cell counting.
RESULTS:
Beta-amyloid plaques, aberrantly phosphorylated tau and neurofibrillary tangles are only found in older patients. The hyperphosphorylated tau tangles are confined to dysplastic neurons. Immunoreactivity for 3- and 4-repeat tau was again only detected within regions of FCD in older patients. With increasing age, the dysplastic cortex became hypocellular and a higher proportion of dysplastic neurons exhibited pathological tau phosphorylation.
CONCLUSIONS:
In older patients, FCD appears more susceptible to formation of pathologically phosphorylated tau neurofibrillary tangles than adjacent histologically normal cortex. Our results suggest a novel convergence of pathological neurodevelopment with pathological age-related neurodegeneration.
Biochim Biophys Acta. 2005 Jan 3;1739(2-3):216-23.
Source
Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Avenue, Chicago, IL 60611, USA. l-binder@northwestern.edu
Abstract
Neurofibrillary tangles (NFT) are comprised of the microtubule-associated protein tau, in the form of filamentous aggregates. In addition to the well-known changes in phosphorylation state, tau undergoes multiple truncations and shifts in conformation as it transforms from an unfolded monomer to the structured polymer characteristic of NFT. Truncations at both the amino- and carboxy-termini directly influence the conformation into which the molecule folds, and hence the ability of tau to polymerize into fibrils. Certain of these truncations may be due to cleavage by caspases as part of the apoptotic cascade. In this review, we discuss evidence that strongly suggests that these truncations occur in an orderly pattern and directly influence the ability of tau to polymerize into filaments.
The Tao of Tau

Tangles, Not Plaques, a New Focus in Alzheimer’s Research
By Rich McManus

Mainstream research on Alzheimer’s disease may be giving short shrift to the tangles part of the two-fisted pathology that has for decades been a hallmark of the disease: the development of plaques and neurofibrillary tangles in the brains of AD patients.

It is the latter feature that intrigues researchers such as Dr. Karen Duff, professor of pathology and cell biology at Columbia University’s Taub Institute for Research on Alzheimer’s Disease and the New York State Psychiatric Institute. She presented a Wednesday Afternoon Lecture in Masur Auditorium on June 23.

In a 75-minute talk titled “It Takes Tau to Tangle: Plaques, Tangles and Neurodegenerative Disease,” Duff explained how scientists are “reconciling the contributions of amyloid plaques (or A-beta, a peptide that forms plaques) and tau,” a protein causing tangles.

.
Accumulation of either substance, she said, has been shown in cell and animal models to lead to loss of synapses, degeneration of neurons and memory loss. However the identification of mutations that cause AD in genes contributing to the amyloid pathway has driven the urge to understand how A-beta or amyloid accumulation contributes to the disease and how it can be neutralized to attempt to prevent or reverse the disease.

Today, however, “the field is moving to tau,” which is normally a soluble microtubule-binding protein. In a misfolded conformation, tau turns bad guy, accumulating in brain cells and destroying them. Once tau pathology begins to propagate in certain brain areas, the plunge from mild cognitive impairment to full-blown AD is virtually assured. In addition to AD, at least 30 different dementia, including Parkinson’s disease, involve what Duff called “tauopathy.”

In her studies on AD, she and her colleagues are looking for possible therapeutic targets against pathways that lead to both plaques and tangles forming. They hypothesize that, in the late-onset or sporadic form of AD (as opposed to the hereditary form, which tends to manifest earlier in life), both paths are activated; preventing one pathway may not prevent the devastating effects of the other unless the therapy is started before the disease has really taken hold. In support of this, human clinical trials have shown that immunotherapy to remove plaque amyloid only had limited success in preventing further decline in the patients and patients still died with end-stage AD, which included devastating tangle pathology.

Once tau pathology begins to propagate in certain brain areas, the plunge from mild cognitive impairment to full-blown AD is virtually assured.
Duff said that although several approaches may be effective against both plaque (or A-beta) pathology and tangles, there are not currently enough being tested; strategies against tau tangles especially need to be researched. A potential vulnerability in brain cells includes degradation pathways and autophagy (literally “self-eating”) pathways, which drugs could theoretically target as a way of spurring the removal of damaged organelles or abnormal proteins such as amyloid or tangle-type tau. Her lab’s recent work using a drug called trehalose that targets autophagy reduced levels of the dangerous form of tau in a mouse model of disease; others have shown similar drugs can remove amyloid and even proteins causing Parkinson’s and Huntington’s disease. Targeting autophagy may eventually prove helpful as a therapy for a range of neurodegenerative diseases or diseases such as AD where several different proteins can accumulate in the same brain.

Interestingly, a person can harbor both plaques and tangles and not have any disease at all. “A crucial thing is where they accumulate,” Duff noted. Her most recen t work aims to look at why certain areas of the brain are vulnerable to plaques and tangles and how we can prevent them from spreading to these sensitive areas.
In summary, Duff concluded:
Tangle pathology correlates best with cognitive decline in AD and it does not seem to be prevented in patients with reduced amyloid load following immunotherapy.
Therapeutics against tau (tangles, precursors) are likely to be necessary to reduce cognitive decline/degeneration once the disease has started.
Disease is likely to be well-advanced at the earliest stage currently diagnosed (MCI, or mild cognitive impairment).
Therapeutic approaches that target more than one pathological entity (plaques, tangles, Lewy bodies) may be more efficacious for diseases such as AD that have mixed pathology.
Dr. Karen Duff’s recent lecture focused on the role of tangles in Alzheimer’s disease and on possible therapeutic interventions





Biochem J. 1991 January 1; 273(Pt 1): 127–133.
PMCID: PMC1150212
Tau in Alzheimer neurofibrillary tangles. N- and C-terminal regions are differentially associated with paired helical filaments and the location of a putative abnormal phosphorylation site.
J P Brion, D P Hanger, M T Bruce, A M Couck, J Flament-Durand, and B H Anderton
Laboratoire d'Anatomie Pathologique et de Microscopie Electronique, Université Libre de Bruxelles, Belgium.
Small right arrow pointing to:This article has been cited by other articles in PMC.
Abstract
To investigate the extent to which whole tau proteins, structurally abnormal tau and fragments of tau are incorporated into neurofibrillary tangles in Alzheimer's disease, an immunocytochemical mapping study using a panel of antibodies to several synthetic human tau peptides has been performed. Neurofibrillary tangles were immunolabelled in situ, and paired helical filaments (PHF), the principal structural component of tangles, were immunolabelled after isolation and Pronase treatment. N-Terminal and C-terminal domains of tau were found to be present in tangles in situ. SDS-treated PHF were found to contain most of the C-terminal half of tau and were also labelled by antibodies to ubiquitin. Only some of these PHF were labelled by antisera to tau sequences towards the N-terminus, and this enabled the identification of a region of tau in which proteolytic cleavage may occur. The ultrastructural appearance of the immunolabelling suggested that both the N- and C-terminal domains of tau extend outwards from the axis of PHF. After Pronase treatment. PHF were strongly labelled only by an antiserum to PHF and by the antiserum to the most C-terminal tau synthetic peptide. The latter antiserum also strongly labelled extracellular tangles in situ, whereas these extracellular tangles were poorly labelled by the antisera to the other synthetic peptides. One anti-(tau peptide) serum labelled a population of neurofibrillary tangles in situ only after alkaline phosphatase pretreatment of tissue sections. Our results show that, although peptides along the length of the tau molecule are associated with neurofibrillary tangles in situ, only the C-terminal one-third of the molecule is tightly associated with PHF, since this region of tau is resistant to SDS treatment of PHF. We also report the existence in PHF in situ of a masked tau epitope which is partially unmasked by dephosphorylation. These results are indicative of post-translational changes in tangle-associated tau in degenerating neurons in Alzheimer's disease.
Am J Pathol. 1987 January; 126(1): 81–91.
PMCID: PMC1899538
Alzheimer's neurofibrillary tangles contain unique epitopes and epitopes in common with the heat-stable microtubule associated proteins tau and MAP2.
S. H. Yen, D. W. Dickson, A. Crowe, M. Butler, and M. L. Shelanski
Ten monoclonal antibodies raised against Alzheimer's neurofibrillary tangles (ANTs) were characterized for reactivity with heat-stable microtubule fractions from bovine and human brain. Five of the antibodies showed very little reaction, but the other five reacted strongly with heat-stable microtubule associated proteins (MAPs). The proteins recognized by these antibodies have estimated molecular weights similar to those of known heat-stable MAPs, tau (52-68 kd) and MAP2 (200-250 kd). That the proteins are indeed tau and MAP2 is demonstrated by reaction of electroblotted proteins with antibodies raised in mouse and guinea pig against bovine brain tau and MAP2. One anti-ANT antibody reacts only with tau, two bind strongly to tau and weakly to MAP2, one recognizes both tau and MAP2 equally well, and one primarily stains MAP2. Extraction of ANT with 2% SDS does not remove tau or MAP2 epitopes from ANT, indicating that epitopes shared with heat-stable MAPs are integral components of ANT. The existence of tau epitopes in ANT is also demonstrated by immunoblotting of ANT-enriched fractions with anti-tau antibodies. Most of the material recognized by anti-tau antibodies in ANT-enriched fractions is present in large molecules excluded by 3% polyacrylamide gel upon electrophoresis. Anti-tau antibodies immunostain ANT in immunofluorescence and immunoperoxidase studies. The immunostaining can be blocked by absorption of anti-tau antibodies with purified tau proteins from bovine brain. Not all ANTs in any given tissue section or isolated Alzheimer perikarial preparations, however, are stained by anti-tau antibodies. These results are consistent with previous studies that have demonstrated heterogeneity of ANTs. Whether this heterogeneity is due to biochemical modification of MAPs or absence of MAPs in some ANTs is unknown. The significance of what appear to be shared epitopes recognized by monoclonal antibodies in tau and MAP2, and the implications this may have on the pathogenesis of ANT formation, requires further investigation.

Reduction of Soluble Aβ and Tau, but Not Soluble Aβ Alone, Ameliorates Cognitive Decline in Transgenic Mice with Plaques and Tangles*

+Author Affiliations
Departments of Neurobiology and Behavior and §Neurology, and Institute for Brain Aging and Dementia, University of California, Irvine, California 92697
1 To whom correspondence should be addressed: Dept. of Neurobiology and Behavior, University of California, Irvine, 1109 Gillespie Neuroscience Bldg. Irvine, CA 92697-4545. Tel.:             949-824-1232 begin_of_the_skype_highlighting            949-824-1232      end_of_the_skype_highlighting      ; Fax: 949-824-7356; E-mail:laferla@uci.edu.

Abstract

Increasing evidence points to soluble assemblies of aggregating proteins as a major mediator of neuronal and synaptic dysfunction. In Alzheimer disease (AD), soluble amyloid-β (Aβ) appears to be a key factor in inducing synaptic and cognitive abnormalities. Here we report the novel finding that soluble tau also plays a role in the cognitive decline in the presence of concomitant Aβ pathology. We describe improved cognitive function following a reduction in both soluble Aβ and tau levels after active or passive immunization in advanced aged 3xTg-AD mice that contain both amyloid plaques and neurofibrillary tangles (NFTs). Notably, reducing soluble Aβ alone did not improve the cognitive phenotype in mice with plaques and NFTs. Our results show that Aβ immunotherapy reduces soluble tau and ameliorates behavioral deficit in old transgenic mice.

Footnotes

2 The abbreviations used are: AD, Alzheimer disease; Aβ, amyloid-β; NFT, neurofibrillary tangles; PBS, phosphate-buffered saline; ELISA, enzyme-linked immunosorbent assay; IL, interleukin; TNF, tumor necrosis factor.
3 L. M. Billings, K. N. Green, J. L. McGaugh, and F. M. LaFerla, submitted manuscript.
4 S. Oddo, A. Caccamo, and F. M. LaFerla, submitted manuscript.
* This work was supported in part by Grant AG0212982 (to F. M. L.) and Grant AG20241 (to D. H. C.) from the NIA, National Institutes of Health and Grant IIRG-02-3767 from the Alzheimer's Association. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

The American Society for Biochemistry and Molecular Biology, Inc.
Biochem Soc Symp. 2001;(67):81-8.

Neurofibrillary tangles and tau phosphorylation.

Source

Laboratory of Histology, Neuroanatomy and Neuropathology, Université Libre de Bruxelles, 808 Route de Lennik, 1070 Brussels, Belgium.

Abstract

Neurofibrillary tangles (NFTs) are a characteristic neuropathological lesion of Alzheimer's disease (AD). They are composed of a highly-phosphorylated form of the microtubule-associated protein tau. We are investigating the relationship between NFTs and microtubule stability and how tau phosphorylation and function is affected in transgenic models and by co-expression with beta-amyloid precursor protein and presenilins. In most NFT-bearing neurons, we observed a strong reduction in acetylated alpha-tubulin immunoreactivity (a marker of stable microtubules) and a reduction of the in situ hybridization signal for tubulin mRNA. In transfected cells, mutated tau forms (corresponding to tau mutations identified in familial forms of frontotemporal dementias linked to chromosome 17) were less efficient in their ability to sustain microtubule growth. These observations are consistent with the hypothesis that destabilization of the microtubule network is an important mechanism of cell dysfunction in Alzheimer's disease. The glycogen synthase kinase-3 beta (GSK-3 beta) generates many phosphorylated sites on tau. We performed a neuroanatomical study of GSK-3 beta distribution showing that developmental evolution of GSK-3 beta compartmentalization in neurons paralleled that of phosphorylated tau. Studies on transfected cells and on cultured neurons showed that GSK-3 beta activity controls tau phosphorylation and tau functional interaction with microtubules. Tau phosphorylation was not affected in neurons overexpressing beta-amyloid precursor protein. Transgenic mice expressing a human tau isoform and double transgenic animals for tau and mutated presenilin 1 have been generated; a somatodendritic accumulation of phosphorylated transgenic tau proteins, as observed in the pretangle stage in AD, has been observed but NFTs were not found, suggesting that additional factors might be necessary to induce their formation.



MARKing tau for tangles and toxicity
http://www.sciencedirect.com/scidirimg/pdf_icon.gif

Gerard DrewesE-mail The Corresponding Author
Cellzome AG, Meyerhofstrasse 1, D-69117 Heidelberg, Germany

Available online 11 September 2004.

In healthy neurons, tau proteins regulate microtubule function in the axon. In the brains of individuals with Alzheimer's disease, tau is hyperphosphorylated and aggregated into intraneuronal deposits called neurofibrillary tangles (NFTs). Hyperphosporylation dislodges tau from the microtubule surface, potentially resulting in compromised axonal integrity and the accumulation of toxic tau peptides. Recent biochemical and animal model studies have re-evaluated tau phosphorylation and other aspects of neurofibrillar pathology. The results indicate that phosphorylation of tau's microtubule-binding domain by the protein kinase MARK primes tau for hyperphosphorylation by the kinases GSK-3 and Cdk5, which in turn triggers the aggregation of tau into filaments and tangles. Toxic consequences for the neuron might be exacerbated by tangle formation but are already evident during the early steps of the process.

Article Outline

Axonopathy, tau abnormalities, and dyskinesia, but no neurofibrillary tangles in p25-transgenic mice
Feng Bian1,*,
Rathna Nath1,
Gregg Sobocinski2,
Robert N. Booher3,
William J. Lipinski1,
Michael J. Callahan1,
Amy Pack1,
Kevin K.-W. Wang1,
Lary C. Walker1
Copyright © 2002 Wiley-Liss, Inc.

Keywords:

Alzheimer;
axon;
cdk5;
electron microscopy;
kinase;
tau;
transgenic;
phosphorylation;
p35;
neurofibrillary tangle;
platelet-derived growth factor

Abstract

Neurofibrillary tangles, one of the pathologic hallmarks of Alzheimer's disease (AD), are composed of abnormally polymerized tau protein. The hyperphosphorylation of tau alters its normal cellular function and is thought to promote the formation of neurofibrillary tangles. Growing evidence suggests that cyclin-dependent kinase 5 (cdk5) plays a role in tau phosphorylation, but the function of the enzyme in tangle formation remains uncertain. In AD, cdk5 is constitutively activated by p25, a highly stable, 25kD protein thought to be increased in the AD brain. To test the hypothesis that p25/cdk5 interactions promote neurofibrillary pathology, we created transgenic mouse lines that overexpress the human p25 protein specifically in neurons. Mice with high transgenic p25 expression have augmented cdk5 activity and develop severe hindlimb semiparalysis and mild forelimb dyskinesia beginning at approximately 3 months of age. Immunohistochemical and ultrastructural analyses showed widespread axonal degeneration with focal accumulation of tau in various regions of the brain and, to a lesser extent, the spinal cord. However, there was no evidence of neurofibrillary tangles in neuronal somata or axons, nor were paired helical filaments evident ultrastructurally. These studies confirm that p25 overexpression can lead to tau abnormalities and axonal degeneration in vivo but do not support the hypothesis that p25-related induction of cdk5 is a primary event in the genesis of neurofibrillary tangles. J. Comp. Neurol. 446:257–266, 2002. © 2002 Wiley-Liss, Inc.

Minireview

Tau Phosphorylation, Tangles, and Neurodegeneration: The Chicken or the Egg?
Daniel H GeschwindCorresponding Author Contact Information, a, E-mail The Corresponding Author
aNeurogenetics Program, Neurology Department, Center for Neurobehavioral Genetics, The Neuropsychiatric Institute, David Geffen School of Medicine, University of California, Los Angeles, 710 Westwood Plaza, Los Angeles, CA 90095 USA

Available online 27 November 2003.

Abstract

Pathological aggregation of the microtubule-associated protein tau is a common feature of many neurodegenerative diseases. Although tau aggregation is associated with abnormal tau phosphorylation, the role of phosphorylation in the initiation of neurodegeneration has been unclear. Now, several animal models and data from human patients provide converging evidence that aberrant tau phosphorylation can cause a neurodegenerative phenotype similar to that seen in human neurodegenerative diseases.

Article Outline

Main Text

Introduction

Pathologic deposition of the microtubule (MT)-associated protein tau, in the form of hyperphosphorylated inclusions or filamentous neurofibrillary tangles (NFTs), is one of the defining features of adult-onset neurodegenerative diseases such as Alzheimer's disease (AD), progressive supranuclear palsy (PSP), and frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) (reviewed in [Lee et al. 2001] and [Buee et al. 2000]). Until recently, much of the genetic evidence for AD pointed to the generation of amyloid-forming Aβ peptides. Hyperphosphorylated insoluble tau in the form of NFTs was considered by many to be a relative bystander, despite the persistent association of tau deposition with dysfunctional neurons within diseased brains and cognitive dysfunction in normal aging (e.g., [Braak and Braak 1996] and[Green et al. 2000]).
The identification of highly penetrant, dominantly inherited mutations in tau causing the related dementia, FTDP-17, demonstrated conclusively that disruption of tau homeostasis could be a primary cause of neurodegeneration (Poorkaj et al. 1998) and (Hutton et al. 1998). FTDP-17 is a clinically and pathologically heterogeneous group of disorders that includes Pick's disease (Lee et al. 2001) and (Bird et al. 2003). A key feature of these conditions is an absence of significant amyloid pathology. However, while tau deposition in the form of NFTs is a defining feature of AD, no mutations in tau have been identified in AD or sporadic cases of FTD, and tau mutations account for only about 15% of inherited FTD cases (Bird et al. 2003) and (Lee et al. 2001). Therefore, understanding the factors that lead to the formation of pathological aggregates in the context of nonmutant tau in AD, FTD, and related neurodegenerative disorders is of major importance. Additionally, in AD, where both insoluble amyloid and tau deposition are present, the temporal or causal relationship between the cascade of amyloid deposition or tau aggregation and the relationship of both of these processes to cell death and dysfunction remain as gaping holes in our understanding.
NFTs seen in patients' brains are intracellular deposits composed of insoluble, hyperphosphorylated tau in a filamentous form, classically the paired helical (PHF), ribbon-like, or straight filament. Normally, tau is distributed extensively and MT associated in axons, but when hyperphosphorylated in its pretangle form, its affinity for MTs is significantly reduced and it becomes preferentially located in the somatodendritic compartment. Tau self-aggregates in vitro in a concentration-dependent manner, forming both straight and paired helical filaments identical to those observed in vivo. This process of tau polymerization is accelerated by a number of factors including polyanionic compounds such as glycosaminoglycans and nucleic acids, certain fatty acids, and hyperphosphorylation (Lee et al. 2001) and (Buee et al. 2000).
Tau phosporylation plays a normal physiologic role in decreasing tau's affinity for MTs and is an important regulator of MT polymerization during development. Tau has approximately 20 to 30 potential phosphorylation sites, many of which are putative targets of proline-directed serine/threonine kinases. Accumulating evidence suggests that two such kinases, glycogen synthase kinase-3β (GSK-3β) and cyclin-dependent kinase-5 (cdk5), are major tau kinases in vitro and in vivo. In AD, FTD, and PSP, insoluble PHF tau and its other pathologically aggregated forms invariably contain tau hyperphosphorylated on residues that overlap with GSK-3β and cdk5 targets. However, until recently it has been unclear whether aberrant tau phosphorylation by these or other kinases, such as p42/44 MAP Kinase (MAPK) and p38 MAPK (Buee et al. 2000) and (Lee et al. 2001), can cause NFT formation in vivo.

Tau Hyperphosphorylation Causes or Enhances NFT Formation

Evidence from the fly (Wittmann et al. 2001) and (Jackson et al. 2002) and the mouse (Cruz et al., 2003 [this issue of Neuron]) suggests that insoluble aggregates such as NFTs and amyloid plaques are signposts of damage already done; tau-related neurodegeneration can occur without, or precede, frank NFT formation. However, processes that accelerate NFT formation inevitably worsen neurodegeneration, supporting the concept that NFTs mark a critical, albeit probably later, process in disease progression. Previous work has shown that tau hyperphosphorylation by GSK-3β in the fly (Jackson et al., 2002) and cdk5 in mouse (Noble et al., 2003) can cause or accelerate NFT formation in vivo, with an attendant worsening of neurodegeneration. However, these effects were observed on a background of either wt tau overexpression or expression of a mutant tau transgene, raising the question of whether significant tau hyperphosphorylation alone under normal conditions may lead to neurofibrillary tau pathology. If so, perhaps tau kinases become an even more attractive target for therapeutic development. Two previous studies of cdk5 dysregulation have shown pathologic tau phosphorylation at the pretangle stage and evidence of disruption of the axonal cytoskeleton. But, while previous studies of p35-25-induced cdk5 overactivity had demonstrated that p25-induced cdk5 activation caused neurodegeneration in vitro (Patrick et al., 1999), no evidence of NFTs or neurodegeneration was identified in these two mouse models of p25/cdk5 overexpression(Ahlijanian et al. 2000) and (Bian et al. 2002).
In this issue of Neuron, Cruz et al. (2003) again examine whether cdk5 activation can cause neurofibrillary pathology and neurodegeneration by overexpressing the cdk5 activator, p25, in the postnatal mouse forebrain under control of an inducible CamK-II promoter, resulting in an approximately 2-fold induction in cdk5 activity and increased specificity for pathological substrates, such as tau and APP. Between 5 and 12 weeks after cdk5 induction, a time-dependent decrease in brain weight, as well as prominent neuronal loss in the cerebral cortex, along with astrogliosis and caspase 3 activation, was observed. Although there was no overt change in tau protein expression itself, insoluble pathologic tau with epitopes and conformation similar to that found in human AD brains was observed to accumulate in the mice overexpressing p25, demonstrated by a combination of immunoblotting, mass spectroscopy, and electron microscopy. Despite the clear biochemical and immunocytochemical evidence of tau hyperphosphorylation followed by its subsequent insolubility and aggregation, no clear neurofibrillary pathology as evidenced by light microscopy, thioflavin-S, or silver staining was seen at 12 to 16 weeks. However, by 27 weeks of cdk5 induction, neurofibrillary-like pathology was abundantly visible on light and electron microscopy in the cerebral cortex and hippocampus. The ultrastructural characteristics of these NFTs and their periodicity have not been characterized, so whether they are orthologous to straight filaments or PHF observed in human FTD or AD is unknown. The interpretation of filament ultrastructure is also complicated by differences in tau isoforms between mice and humans, which could lead to different filament morphologies. However, the phosphorylation pattern, insolubility, and light microscopic appearance described by Cruz et al. are consistent with these deposits being typical NFTs, and their distribution correlates with the regional pattern of cdk5 activation. In the future, it will be important to determine filament ultrastructure and the relationship between behavioral abnormalities and the timing and topology of neuronal loss and NFT appearance.
Why this model of cdk5 activation produces striking neurodegeneration and NFT formation, and previous models did not, is not known. Cruz et al. suggest that NFT formation is most likely due to the significantly greater cdk5 activation present in this model relative to previous models. The differences in promoters used in each of these models may also be contributory. In any regard, Cruz et al. provide the strongest evidence to date that aberrant tau kinase activity of any kind can lead to neurodegeneration and tau neurofibrillary pathology in vivo in the absence of tau dysregulation or mutations. These findings add to a growing body of evidence that NFTs or NFT-like pathology can be caused and/or accelerated by tau kinase overactivity in vivo (e.g., [Jackson et al. 2002], [Noble et al. 2003] and [Liou et al. 2003]) and firmly establishes tau phosphorylation as a potential therapeutic target in AD and FTD.
Recently, Noble et al. (2003) demonstrated that p25/cdk5 overactivation, in conjunction with expression of human tau containing the most common FTD-causing mutation in mouse, is associated with concomitant GSK-3α/β activation. Thus, although p25/cdk5 activation provides the initial insult in the Noble et al. model, GSK-3β is likely to be playing a role. Why GSK-3β activation is not observed in the model developed by Cruz et al. is not known—it may not be relevant in the context of such high cdk5 activity. The authors propose that GSK-3β may act to produce NFTs in the context of mutant or overexpressed tau, rather than normal tau. However, the persistent localization of GSK-3β with pretangle and tangle-bearing neurons in human AD suggests otherwise.
Although the circumstantial evidence is strong, it is not yet certain whether the neurodegeneration in the Cruz et al. model of p25 overexpression is a direct consequence of tau hyperphosphorylation by cdk5 or occurs via another signaling cascade. Cdk5 has many substrates, including proteins associated with neurodegeneration, such as APP (which are preferentially hyperphosphorylated by the p25/cdk5 complex; [Cruz et al. 2003] and [Dhavan and Tsai 2001]). Cdk5 is also involved in several aspects of neuronal differentiation as a link between extracellular signals and the cytoskeleton and thus could lead to cell dysfunction and death via a number of different pathways. Additionally, Cruz et al. demonstrate that tau is hyperphosphorylated on sites that are not known cdk5 targets and that two other known tau kinases may be activated, suggesting that at least some of the effects of p25 overactivation are indirect. Whether this indirect effect occurs through activation of other kinases, by downregulation of phosphatases, or somehow by altering the affinity of peptidyl-prolyl isomerases for tau (favoring a conformation not permissive for tau dephosphorylation) are important issues that now need to be addressed. Nevertheless, it is becoming clear that direct or indirect tau hyperphosphorylation via at least two proline-directed kinases can accelerate or cause neurodegeneration and NFT formation.
The importance of dephosphorylation in tauopathy is highlighted by the ability of the phosphorylation-dependent prolyl isomerase, Pin1, to provide relative protection from age-dependent neurodegeneration (Liou et al., 2003). Pin1 recognizes specific phosphorylated serine or threonine residues in tau that are followed by proline residues and catalyzes a critical conformational change that allows dephosphorylation at these residues to occur. Pin1 expression is inversely correlated with markers of neurofibrillary pathology in human AD brains, and Pin−/−mice develop tau hyperphosphorylation, NFTs, and age-dependent neurodegeneration (Liou et al., 2003). This pathology is very similar to that caused by misexpression of either mutant or wt tau in other models (e.g., [Lewis et al. 2001], [Allen et al. 2002] and [Andorfer et al. 2003]), emphasizing that multiple pathways affecting tau expression levels, dephosphorylation (e.g., Kins et al., 2001), and phosphorylation may coalesce in a common phenotype characterized by tau hyperphosphorylation, aggregation, and NFT formation. Current evidence shows that the known FTDP-17-causing tau mutations either disrupt MT binding or affect tau splicing, leading to tau isoform imbalance (Lee et al. 2001) and (Hutton et al. 1998). Therefore, one feature potentially in common with the human mutations and animal models of tauopathy is an increase in free tau unbound to microtubules. Since tau polymerization into filaments is concentration dependent in vitro, this provides an obvious common mechanism for tau aggregation and NFT formation in vivo(Figure 1).



Full-size image (40K) - Opens new window
Figure 1. Tau Aggregation and Neurodegeneration: The Balance of Tau Phosphorylation
Tau is tightly balanced between being MT bound or free, depending on the state of the cell. Factors that promote an increase in unbound tau and aggregation are highlighted with red arrows (e.g., tau kinases), and protective factors are blue (e.g., tau phosphatases). This model focuses on a toxic gain-of-function mechanism based on increased tau aggregation and depicts cdk5 as acting upstream of GSK-3β. Loss of function, such as cytoskeletal disruption based on tau hyperphosphorylation and MT dysregulation, may contribute as well. Genetic and environmental factors may act at many steps. For example, tau mutations lead to increased free tau or tau isoforms that may be more prone to aggregate, whereas ApoE may act on Aβ42 and Tau. For many of the tauopathies, such as FTD, this process occurs independently of Aβ42.


This of course begs several questions: is there a common mechanism that leads to neurodegeneration in these different models of tauopathy, whether the model is based on tau misexpression, mutation, or altered kinase activity? The pervasive association of tau insolubility, the common pattern of pathological tau phosphorylation, and the histological signs of tau deposition observed in these models suggest that this is the case, but it is not certain. Perhaps, most relevant to the current discussion, are cdk5, GSK-3β, and other tau kinases acting in series or in parallel? The recent paper by Noble et al. (2003) and in vitro evidence suggest that cdk5 could be upstream of GSK-3β, serving to prime tau at key epitopes that modulate tau's affinity for microtubules. This hypothesis can be tested in vivo, since the detrimental effects of GSK-3β should be attenuated on a background of conditional cdk5 inactivation. Furthermore, specific kinase inhibitors can be tested to assess their ability to modulate the pathology observed in animals with mutant tau transgenes to test the primacy of particular kinase or phosphatase pathways. Of course, all of these mechanisms being studied in animal models need to be considered from the perspective of their potential overlap with the pathological alterations observed in human subjects (e.g., [Patrick et al. 1999] and [Liou et al. 2003]).

Tau, Amyloid, and Regional Vulnerability

Finally, it is necessary to consider tau phosphorylation in the context of two issues that seem central to our understanding and treatment of these neurodegenerative diseases. (1) How might these pathways involving tau intersect with the amyloid cascade that is proposed to lead to neurodegeneration in AD? (2) Do any of these processes or mechanisms explain the regional vulnerability that is the hallmark of human neurodegenerative diseases, such as FTD and AD? With regard to the first question, current evidence supports that view that Aβ could lie upstream of tau in AD, since increased Aβ42 leads to increased neurodegeneration and NFTs in mouse models carrying FTDP-17 causing tau mutations. We have previously speculated that GSK-3β could potentially mediate this toxic effect of amyloid (Jackson et al., 2002). Fibrillar Aβ42 fragments induce GSK-3β activation in vitro, leading to tau phosphorylation and apoptosis. Other kinases such as p38 MAPK, p42/44 MAPK, and CDK5 may play similar roles (Lee et al. 2000) and(Zheng et al. 2002). This cascade of kinase induction by Aβ42 could be a particularly vicious circle, as at least one form of GSK3 is involved in Aβ42 production (Phiel et al., 2003). In the case of sporadic disease, it is also equally plausible that genetic and environmental factors that increase amyloid deposition, such as ApoE isoforms, oxidative stress, etc., also induce tau aggregation or hyperphosphorylation, and the two aggregation pathways are initiated concurrently for AD, or alone, as would be the case in FTD.
One of the most constant features of AD and FTD are their distinct clinical presentations, which reflect the distinct early regional pathology in these disorders. We have speculated that this regional vulnerability in FTD may have a developmental component that alters neuronal patterning or phenotype generation in a manner that would render that region more susceptible to the later insults of aging (Geschwind and Miller, 2001). Genes with a dual role in patterning and neurodegeneration could contribute to the focal nature of these disorders, and both cdk5 and GSK3 have important roles in CNS patterning and development outside of their role as tau kinases (Anderton et al., 2000).
Recent evidence indicates that the regulation of tau phosphorylation is another potential source of regional susceptibility. For example, cdk5 overactivation under the control of the NSE promoter shifts the distribution of pathology in transgenic mice overexpressing the P301L mutation to cortical, in addition to more subcortical regions (Noble et al., 2003). The interpretation of regional vulnerability in animal models is confounded by the effect of different promoters, genetic background, and transgene insertion position. Although the use of a relatively ubiquitous neuronal promoter lessens these concerns, it is still important to consider the human data when assessing regional vulnerability. In human AD, the distribution of both GSK-3β and cdk5 are associated with pretangle or tangle-bearing neurons, suggesting that both neurons could be involved in early vulnerability (Patrick et al. 1999), (Pei et al. 1999) and (Yamaguchi et al. 1996). In this regard, it is also notable that Pin1 expression is highest in human neuronal populations that are less vulnerable to AD, consistent with its neuroprotective function (Liou et al., 2003). However, the more superficial layers of cortex where Pin1 was reported to be the most abundant contain the most vulnerable populations in FTD. This emphasizes that the individual factors leading to regional vulnerability in one dementia may not be acting in another, since the primary regions affected are so distinct. In any event, the concept of regional vulnerability represents something of a paradox, since the known disease-causing mutations all affect proteins that are widely expressed. Resolving this paradox, and therefore understanding the mechanisms underlying this focal susceptibility, is central to understanding these neurodegenerative diseases.

Acknowledgements

The author thanks his collaborators and coworkers, especially George Jackson, M.D., Ph.D., and Martina Wiedau-Pazos, M.D., Ph.D., for stimulating discussions and Bonita Porch for editorial assistance. Because of space limitations, the author apologizes for not being able to cite all who have made significant contributions to this growing body of knowledge.

References

Ahlijanian et al. 2000 M.K Ahlijanian, N.X Barrezueta, R.D Williams, A Jakowski, K.P Kowsz, S McCarthy, T Coskran, A Carlo, P.A Seymour and J.E Burkhardt et al., Proc. Natl. Acad. Sci. USA 97(2000), pp. 2910–2915. View Record in Scopus | Cited By in Scopus (201)
Allen et al. 2002 B Allen, E Ingram, M Takao, M.J Smith, R Jakes, K Virdee, H Yoshida, M Holzer, M Craxton and P.C Emson et al., J. Neurosci. 22 (2002), pp. 9340–9351. View Record in Scopus |Cited By in Scopus (175)
Anderton et al. 2000 B.H Anderton, R Dayanandan, R Killick and S Lovestone, Mol. Med. Today 6(2000), pp. 54–59. Abstract | Article | http://www.sciencedirect.com/scidirimg/icon_pdf.gif PDF (102 K) | View Record in Scopus | Cited By in Scopus (39)
Andorfer et al. 2003 C Andorfer, Y Kress, M Espinoza, R de Silva, K.L Tucker, Y.A Barde, K Duff and P Davies, J. Neurochem. 86 (2003), pp. 582–590. View Record in Scopus | Cited By in Scopus (150)
Bian et al. 2002 F Bian, R Nath, G Sobocinski, R.N Booher, W.J Lipinski, M.J Callahan, A Pack, K.K Wang and L.C Walker, J. Comp. Neurol. 446 (2002), pp. 257–266. View Record in Scopus |Cited By in Scopus (66)
Bird et al. 2003 T Bird, D Knopman, J VanSwieten, S Rosso, H Feldman, H Tanabe, N Graff-Raford, D Geschwind, P Verpillat and M Hutton, Ann. Neurol. 54 (Suppl 5) (2003), pp. S29–S31.View Record in Scopus | Cited By in Scopus (44)
Braak and Braak 1996 H Braak and E Braak, Acta Neurol, Scand. Suppl. 165 (1996), pp. 3–12.View Record in Scopus | Cited By in Scopus (243)
Buee et al. 2000 L Buee, T Bussiere, V Buee-Scherrer, A Delacourte and P.R Hof, Brain Res. Rev. 33 (2000), pp. 95–130. Abstract | Article | http://www.sciencedirect.com/scidirimg/icon_pdf.gif PDF (2399 K) | View Record in Scopus | Cited By in Scopus (621)
Cruz et al. 2003 J.C Cruz, H.-C Tseng, J.A Goldman, H Shih and L.-H Tsai, Neuron 40 (2003), pp. 471–483 , this issue. Article | http://www.sciencedirect.com/scidirimg/icon_pdf.gif PDF (711 K) | View Record in Scopus | Cited By in Scopus (204)
Dhavan and Tsai 2001 R Dhavan and L.H Tsai, Nat. Rev. Mol. Cell Biol. 2 (2001), pp. 749–759.View Record in Scopus | Cited By in Scopus (459)
Geschwind and Miller 2001 D.H Geschwind and B.L Miller, Am. J. Med. Genet. 101 (2001), pp. 370–381. View Record in Scopus | Cited By in Scopus (50)
Green et al. 2000 M.S Green, J.A Kaye and M.J Ball, Neurology 54 (2000), pp. 105–113. View Record in Scopus | Cited By in Scopus (63)
Hutton et al. 1998 M Hutton, C.L Lendon, P Rizzu, M Baker, S Froelich, H Houlden, S Pickering-Brown, S Chakraverty, A Isaacs and A Grover et al., Nature 393 (1998), pp. 702–705.
Jackson et al. 2002 G.R Jackson, M Wiedau-Pazos, T.K Sang, N Wagle, C.A Brown, S Massachi and D.H Geschwind, Neuron 34 (2002), pp. 509–519. Article | http://www.sciencedirect.com/scidirimg/icon_pdf.gif PDF (581 K) | View Record in Scopus | Cited By in Scopus (232)
Kins et al. 2001 S Kins, A Crameri, D.R Evans, B.A Hemmings, R.M Nitsch and J Gotz, J. Biol. Chem. 276 (2001), pp. 38193–38200. View Record in Scopus | Cited By in Scopus (101)
Lee et al. 2000 M.S Lee, Y.T Kwon, M Li, J Peng, R.M Friedlander and L.H Tsai, Nature 405(2000), pp. 360–364. View Record in Scopus | Cited By in Scopus (547)
Lee et al. 2001 V.M Lee, M Goedert and J.Q Trojanowski, Annu. Rev. Neurosci. 24 (2001), pp. 1121–1159. View Record in Scopus | Cited By in Scopus (898)
Lewis et al. 2001 J Lewis, D.W Dickson, W.L Lin, L Chisholm, A Corral, G Jones, S.H Yen, N Sahara, L Skipper and D Yager et al., Science 293 (2001), pp. 1487–1491. View Record in Scopus | Cited By in Scopus (619)
Liou et al. 2003 Y.C Liou, A Sun, A Ryo, X.Z Zhou, Z.X Yu, H.K Huang, T Uchida, R Bronson, G Bing and X Li et al., Nature 424 (2003), pp. 556–561. View Record in Scopus | Cited By in Scopus (152)
Noble et al. 2003 W Noble, V Olm, K Takata, E Casey, O Mary, J Meyerson, K Gaynor, J LaFrancois, L Wang and T Kondo et al., Neuron 38 (2003), pp. 555–565. Article | http://www.sciencedirect.com/scidirimg/icon_pdf.gif PDF (454 K) | View Record in Scopus | Cited By in Scopus (188)
Patrick et al. 1999 G.N Patrick, L Zukerberg, M Nikolic, S de la Monte, P Dikkes and L.H Tsai,Nature 402 (1999), pp. 588–589.
Pei et al. 1999 J.J Pei, E Braak, H Braak, I Grundke-Iqbal, K Iqbal, B Winblad and R.F Cowburn, J. Neuropathol. Exp. Neurol. 58 (1999), pp. 1010–1019. View Record in Scopus | Cited By in Scopus (234)
Phiel et al. 2003 C.J Phiel, C.A Wilson, V.M Lee and P.S Klein, Nature 423 (2003), pp. 435–439.View Record in Scopus | Cited By in Scopus (491)
Poorkaj et al. 1998 P Poorkaj, T.D Bird, E Wijsman, E Nemens, R.M Garruto, L Anderson, A Andreadis, W.C Wiederholt, M Raskind and G.D Schellenberg, Ann. Neurol. 43 (1998), pp. 815–825. View Record in Scopus | Cited By in Scopus (731)
Wittmann et al. 2001 C.W Wittmann, M.F Wszolek, J.M Shulman, P.M Salvaterra, J Lewis, M Hutton and M.B Feany, Science 293 (2001), pp. 711–714. View Record in Scopus | Cited By in Scopus (314)
Yamaguchi et al. 1996 H Yamaguchi, K Ishiguro, T Uchida, A Takashima, C.A Lemere and K Imahori, Acta Neuropathol. (Berl.) 92 (1996), pp. 232–241. View Record in Scopus | Cited By in Scopus (147)
Zheng et al. 2002 W.H Zheng, S Bastianetto, F Mennicken, W Ma and S Kar, Neuroscience 115(2002), pp. 201–211. Abstract | Article | http://www.sciencedirect.com/scidirimg/icon_pdf.gif PDF (493 K) | View Record in Scopus | Cited By in Scopus (68)

Correspondence: Daniel H. Geschwind,             310-206-6814 begin_of_the_skype_highlighting            310-206-6814      end_of_the_skype_highlighting       (phone), 310-267-2401 (fax)
Volume 88, Number 2, 113-121, DOI: 10.1007/BF00294503

Corticobasal degeneration: a disease with widespread appearance of abnormal tau and neurofibrillary tangles, and its relation to progressive supranuclear palsy

Abstract

The neuropathological findings, including immunohistochemistry and electron microscopy, of two patients with clinical findings consistent with corticobasal degeneration (CBD) are reported. Both patients showed degeneration of the precentral cortex, the substantia nigra, the pallidum, and the thalamus. Many ballooned neurons were seen in the cerebral cortex, and argentophilic, skein-like inclusions suggesting neurofibrillary tangles (NFTs) were found in the brain stem and precentral cortex in patient 1. In contrast, patient 2 clearly showed NFTs in the brain stem and dentate nucleus which were indistinguishable from those seen in progressive supranuclear palsy (PSP), while only a few ballooned neurons were found in the cerebral cortex. Gallyas silver stain showed many argentophilic inclusions suggesting NFTs in the brain stem, subcortical nuclei, and cerebral cortex in both patients. Immunohistochemistry for tau showed tau-positive neurons in the cerebral cortex, brain stem, subcortical nuclei and spinal cord, and tau-positive glial cells were seen in the cerebral cortex, white matter and subcortical nuclei, and thread-like structures were seen in the cerebral cortex and white matter. Electron microscopy of the brain stem showed NFTs consisting of paired helical filaments in patient 1, and paired helical filaments and straight tubules in patient 2. Immunoelectron microscopy revealed parallel tau-positive filaments in the cerebral cortex in patent 1. From the two patients, the widespread appearance of abnormal tau and NFTs is one of the essential pathological features in CBD, and it also appears that CBD and PSP have some common underlying pathological processes. Patient 2 is closer to PSP than patient 1 and suggests CBD would link to PSP.
Key words  Corticobasal degeneration - Progressive supranuclear palsy - Neurofibrillary tangles - Abnormal tau

Fulltext Preview

Tau antisera recognize neurofibrillary tangles in a range of neurodegenerative disorders
Dr Catharine L. Joachim MD1,2,*,
James H. Morris BM, BCH, PhD2,
Kenneth S. Kosik MD1,
Dennis J. Selkoe MD1
Article first published online: 8 OCT 2004
DOI: 10.1002/ana.410220411
Copyright © 1987 American Neurological Association

Abstract

Neurofibrillary tangles occur in a number of apparently distinct neurodegenerative diseases and in normal aging of the human brain. Antibodies raised against Alzheimer's disease paired helical filaments immunolabel the tangles seen in all other tangle-associated disorders examined to date. The neuronal microtubule-associated protein, tau, has recently been identified as an antigenic component of neurofibrillary tangels and senile plaque neurites in Alzheimer's disease. Three different polyclonal antibodies with strong tau immunoreactivity are examined in this study. These antibodies were found to immunostain tangles in normal aged brain and in brains affected by a range of neurodegenerative disorders, including Down's syndrome, Alzheimer's disease plus Parkinson's disease, progressive supranuclear palsy, and the parkinsonism-dementia complex of Guam, as well as Pick bodies in Pick's disease. The findings further illustrate the relative nonspecificity of neurofibrillary lesions in neurodegenerative disorders.
      Catharine L. Joachim James H. Morris Kenneth S. Kosik Dennis J. Selkoe
Chronic lithium administration to FTDP-17 tau and GSK-3β overexpressing mice prevents tau hyperphosphorylation and neurofibrillary tangle formation, but pre-formed neurofibrillary tangles do not revert
Tobias Engel,
Paloma Goñi-Oliver,
José J. Lucas,
Jesús Avila,
Félix Hernández
Article first published online: 31 JUL 2006
DOI: 10.1111/j.1471-4159.2006.04139.x

Keywords:

Alzheimer's disease;
frontotemporal dementia and parkinsonim linked to chromosome 17;
glycogen synthase kinase-3;
lithium;
neurofibrillary tangles

Abstract

Glycogen synthase kinase-3 (GSK-3) has been proposed as the main kinase able to aberrantly phosphorylate tau in Alzheimer's disease (AD) and related tauopathies, raising the possibility of designing novel therapeutic interventions for AD based on GSK-3 inhibition. Lithium, a widely used drug for affective disorders, inhibits GSK-3 at therapeutically relevant concentrations. Therefore, it was of great interest to test the possible protective effects of lithium in an AD animal model based on GSK-3 overexpression. We had previously generated a double transgenic model, overexpressing GSK-3β in a conditional manner, using the Tet-off system and tau protein carrying a triple FTDP-17 (frontotemporal dementia and parkinsonism linked to chromosome 17) mutation. This transgenic line shows tau hyperphosphorylation in hippocampal neurones accompanied by neurofibrillary tangles (NFTs). We used this transgenic model to address two issues: first, whether chronic lithium treatment is able to prevent the formation of aberrant tau aggregates that result from the overexpression of FTDP-17 tau and GSK-3β; second, whether lithium is able to change back already formed NFTs in aged animals. Our data suggest that progression of the tauopathy can be prevented by administration of lithium when the first signs of neuropathology appear. Furthermore, it is still possible to partially reverse tau pathology in advanced stages of the disease, although NFT-like structures cannot be changed. The same results were obtained after shut-down of GSK-3β overexpression, supporting the possibility that GSK-3 inhibition is not sufficient to reverse NFT-like aggregates.
Brain Research
Volume 601, Issues 1-2, 22 January 1993, Pages 164-172


Lack of the caryl terminal sequence of tau in ghost tangles of Alzheimer's disease
http://www.sciencedirect.com/scidirimg/pdf_icon.gif

Riuko Endoha, Midori Ogawarab, Takeshi Iwatsuboa, Imaharu Nakanocand Hiroshi MoriCorresponding Author Contact Information, a, b
aDepartment of Neuropathology, Institute of Brain Research, University of Tokyo School of Medicine, Tokyo (Japan)
bDepartment of Neurophysiology, Tokyo Metropolitan Institute of Gerontology, Tokyo (Japan)
cDepartment of Neurophysiology, Tokyo Metropolitan Institute of Neuroscience, Tokyo (Japan)
Accepted 18 August 1992.
Available online 6 March 2003.

Using seven independent antibodies against the amino terminal to the caryl sequence of tau, we biochemically analyzed and compared the neuropathogenesis of two Alzheimer's disease brains from the viewpoint of abnormal processing on tau, the major constituent of paired helical filaments. One showed typical Alzheimer's disease with senile plaques and intracellular neurofibrillary tangles. The other showed advanced Alzheimer's disease with senile plaques and virtually the sole of ghost tangles without intracellular neurofibrillary tangles. We confirmed the previous observation that the caryl thirds of tau are tightly associated with paired helical filaments isolated in the presence of SDS. We found that biochemically, ghost tangles were abnormally phosphorylated and lacked the final caryl terminal sequence as well as the amino half of tau, unlike intracellular tangles. From these biochemical results taken together with the current evidence for ubiquitin in ghost tangles, we concluded that ghost tangles were extensively processed and irreversibly transformed into highly insoluble extracellular deposits.
Keywords: Tau; Ghost tangle; Alzheimer's disease; Processing; Abnormal phosphorylation

Corresponding Author Contact InformationCorrespondence: H. Mori, Department of Neuropathology, Institute of Brain Research, Faculty of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyoku, Tokyo 113, Japan. Fax: (81) (813) 3814-8154.


Ultrastructural localization of beta-amyloid, tau, and ubiquitin epitopes in extracellular neurofibrillary tangles

G Perry, and
+Author Affiliations
Department of Neurology, University of Genoa, Italy.

Abstract

Neurofibrillary tangles (NFTs), a hallmark of Alzheimer disease, are commonly located in perikarya of neurons. In advanced cases of Alzheimer disease, however, NFTs are observed also in the extracellular space. As extracellular NFTs (E-NFTs), and occasionally intracellular NFTs (I-NFTs), are recognized by antibodies to beta-amyloid protein (beta AP), beta AP may be present not only in amyloid deposits but also in paired helical filaments (PHFs), the primary components of NFTs. We compared the antigenic characteristics of I-NFTs and E-NFTs with light- and electron-microscopic immunocytochemistry by using several antibodies to noncontiguous epitopes of the microtubule-associated protein tau and of ubiquitin (Ub) as well as an antiserum to beta AP. At variance with I-NFTs, E-NFTs were made predominantly of straight filaments (SFs), rather than PHFs, that were often separated by astroglial processes and in close association with small beta AP deposits. Occasionally, E-NFTs were made of bundles of amorphous material, which showed no resemblance to SFs, PHFs, or amyloid fibrils. The antigenic changes in E-NFTs suggest that when NFTs become extracellular they lose the N and, possibly, the C termini of tau while maintaining the intermediate region of the molecule; they also lose the N-terminal two-thirds of Ub while the C-terminal conjugation site of Ub is preserved. A small subset of E-NFTs reacted with antibodies to both beta AP and tau. Although in most E-NFTs, the epitopes recognized by tau and Ub antibodies were located in typical PHFs and SFs, the epitopes recognized in this subset of anti-beta AP and anti-tau-positive E-NFTs were located exclusively in the bundles of amorphous material. It is suggested that either beta AP epitopes are present but inaccessible in PHFs and SFs and become exposed after conformational changes occurring in the extracellular space or PHFs and SFs become closely associated with beta AP in the extracellular space.

Original Articles

Glial Tau Pathology in Neurodegenerative Diseases: Their Nature and Comparison with Neuronal Tangles
http://www.sciencedirect.com/scidirimg/pdf_icon.gif


K. IkedaA, *, H. AkiyamaA, T. AraiA and T. NishimuraA
A Department of Neuropathology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo, 156, Japan

Available online 20 August 1998.

Abstract

Tau-positive inclusions that occur in glial cells are called glial fibrillary tangles or, more simply, glial tangles. These include tuft-shaped astrocytes, thorn-shaped astrocytes, coiled bodies, and argyrophilic threads. The latter two structures occur in oligodendroglia. The tau protein in glial tangles is hyperphosphorylated and has similar immunohistochemical profiles to that in neurofibrillary tangles (NFTs) except that there are no epitopes derived from alternatively spliced exon 2 and 3. In contrast to NFTs, glial tangles rarely show solid filaments. Such NFT-associated molecules as ubiquitin, apolipoprotein E, alpha1-antichymotrypsin, and heparan sulfate are all absent from glial tangles. These characteristics suggest that glial tangles resemble the pre-tangles that occur in neurons and are thought to represent an early stage of NFTs. Tau pathology in neurodegenerative diseases takes heterogenous forms.
Author Keywords: Glial tangles; Neurofibrillary tangles; Pre-tangles

Article Outline

Volume 90, Number 6, 620-625, DOI: 10.1007/BF00318575

Thorn-shaped astrocytes: possibly secondarily induced tau-positive glial fibrillary tangles

Abstract

Argyrophilic and tau-positive abnormal structures occurring in glial cells are called glial fibrillary tangles. In the astrocyte, a conspicuous tau-positive structure is known to appear in progressive supranuclear palsy (PSP). In this report, another type of argyrophilic and tau-positive astrocytes is reported. The morphology of this new type is quite different from that of the previously reported tau-positive astrocyte in PSP and they are designated here as thorn-shaped astrocytes (TSA). TSA have an apparently argyrophilic cytoplasm with a few short processes and often have a small eccentric nucleus, whose appearance resembles that of a reactive astrocyte. Immunohistochemically, TSA are positive to anti-tau antibodies but are negative for ubiquitin. Simultaneous immunostaining revealed the coexistence of tau and glial fibrillary acidic protein epitopes in the same cytoplasm. Electron microscopically, bundles of 15-nm straight tubules were included in the cytoplasm together with abundant glial filaments. In the vicinity of a cluster of TSA, related structures of perivascular or subpial tau-positive linings, which correspond to astrocytic end-feet, are sometimes observed. In almost all cases, a few TSA are generally located in a confined area of subpial and subependymal regions. Although TSA appear to be intimately associated with some diseases, they are also found in a wide range of cytoskeletal disorders including the aged brain with neurofibrillary tangles. TSA are presumed to be a secondarily induced product in relation to astrocytic reaction.
Key words  Thorn-shaped astrocyte - Glial fibrillary tangles - Tau - Astrocyte - Straight tubules
Neurofibrillary Tangles in Progressive Supranuclear Palsy Contain the Same Tau Epitopes Identified in Alzheimer's Disease PHFtau
Schmidt, Marie Luise PhD; Huang, Richard BS; Martin, John A. BS; Henley, Jewel BS; Mawal-Dewan, Madhumalti PhD; Hurtig, Howard I. MD; -Y. Lee, Virginia M. PhD; Trojanowski, John Q. MD, PhD
Collapse Box
Abstract
Neurofibrillary tangle (NFT)-rich brain samples from patients with progressive supranuclear palsy (PSP) or Alzheimer's disease (AD) were probed with a large panel of anti-tau antibodies to compare the species of tau present in PSP and AD NFTs by immunohistochemistry and Western blot methods. These antibodies have been shown to recognize phosphate-independent or -dependent epitopes that extend from the amino to the carboxy terminal domains of normal brain tau and the abnormal tau in the paired helical filaments (PHFs) of AD NFTs (PHFtau). The immunohistochemical studies showed that all of the tau epitopes detected in brainstem PSP NFTs also were found in hippocampal AD NFTs and vice versa. While Western blots demonstrated 2 PHFtau-like immunobands in PSP brainstem, a triplet of PHFtau proteins were seen in the AD and PSP hippocampus.
Despite differences in the distribution, ultrastructure and immunoblot profile of NFTs in PSP and AD, the same constellation of tau epitopes is present in the abnormal tau proteins in PSP and AD NFTs. Thus, the generation of abnormal tau proteins in PSP (PSPtau) and AD (PHFtau) may have similar adverse biological consequences in both diseases.
(C) 1996 American Association of Neuropathologists, Inc



Abnormally phosphorylated tau protein related to the formation of neurofibrillary tangles and neuropil threads in the cerebral cortex of sheep and goat
http://www.sciencedirect.com/scidirimg/pdf_icon.gif

Heiko BraakCorresponding Author Contact Information, a, Eva Braaka and Martin Strothjohanna
aDepartment of Anatomy, J.W. Goethe University, Theodor Stern Kai 7, 60590 Frankfurt, FRG
Received 31 January 1994;
accepted 14 February 1994.
Available online 5 March 2003.

Abstract

Frontal sections including temporal isocortex, entorhinal region and hippocampus from aged domestic animals (dog, cat, horse, sheep and goat) were studied for Alzheimer-related changes using immunostaining with the AT8 antibody for abnormally phosphorylated tau protein and selective silver techniques for A4 amyloid and neurofibrillary changes of the Alzheimer type. The material available to us did not show A4 amyloid deposits or argyrophilic neurofibrillary changes. Only the brains of aged sheep and goat exhibited the presence of AT8-immunoreactive pyramidal cells in the entorhinal region and hippocampal formation. Two groups of AT8-positive neurons could be observed: The first group contained evenly distributed immunoreactive material in all parts of the soma, the dendrites and the axon. The neuronal processes appeared quite normal. The second group, however, showed conspicuous changes in the cellular processes consisting of a loss of immunoreactivity within the axon and the proximal dendrites and the appearance of intensely stained swellings within the curved distal dendrites. These changes were closely reminiscent to alterations of the cytoskeleton known to occur at the same location in the aging human brain and in Alzheimer's disease. The findings justify a closer look at sheep and goat when searching for suitable animal models for experimental studies of the conditions responsible for the development of Alzheimer-related neurofibrillary changes.
Keywords: Animal model; Alzheimer's disease; Cytoskeleton; Neurofibrillary change; Tau protein; Abnormal phosphorylation

Corresponding Author Contact InformationCorresponding author: Fax: (49) 69-6301-6425.
Volume 84, Number 6, 596-605, DOI: 10.1007/BF00227736

Immunocytochemistry of neurofibrillary tangles with antibodies to subregions of tau protein: identification of hidden and cleaved tau epitopes and a new phosphorylation site

Abstract

Antibodies to multiple epitopes spanning the length of the tau molecule were used to study Alzheimer neurofibrillary tangles (NFT) using immunocytochemical methods and several differnt methods of fixation and tissue processing, including staining of vibratome sections, hydrated autoclaving of paraffin sections and immunofluorescence of NFT isolated from fresh brain tissue. Smears and sections were pretreated with trypsin and/or phosphatase to further characterize antibody binding. In tissue fixed briefly in periodate-lysine-paraformaldehyde, tau immunoreactivity was detected in astrocytes, but only a few tau epitopes were detected in NFT with this fixation method. In contrast, all tau epitopes were detected in NFT in tissue fixed in formaldehyde for prolonged periods of time. In the hippocampus, the number of NFT detected in the dentate fascia was in proportion to the duration of dementia, as we previously noted. Dentate fascia NFT were intracellular (i-NFT) and were reactive with antibodies recognizing epitopes in both the carboxy- and amino-terminal regions of tau, but not the microtubule-binding domain of tau, suggesting that microtubule-binding domain epitopes are hidden in i-NFT. In contrast, NFT in the subiculum and layer II of the parahippocampal cortex were mostly extracellular (e-NFT), especially in severe cases of long duration, e-NFT were immunoreactive with antibodies to the microtubule-binding domain, but only weakly reactive with antibodies to carboxy- or amino-terminal epitopes, suggesting that e-NFT may contain fragments of tau. In both isolated NFT and NFT in sections, amino-terminal epitopes, including the Alz-50 epitope, were sensitive to trypsin proteolysis, which suggests that the lack of staining of e-NFT by antibodies to the amino-terminal regions of tau is due to proteolysis. Antibodies reactive with amino-terminal epitopes also stained fewer NFT following hydrated autoclaving, while those reacting with the carboxy half of tau stained more NFT after hydrated autoclaving. Thus, although carboxy-terminal regions are not detected in e-NFT, they are probably masked, rather than proteolytically cleaved, since they can be revealed by hydrated autoclaving. Finally, phosphatase treatment of isolated NFT revealed enhanced immunostaining not only with Tau-1, as in previous studies demonstrating abnormal phosphorylation of tau proteins in NFT, but also with an antibody to exon 2, which reveals yet another phosphorylation site in tau of NFT.
Key words  Alzheimer's disease - Immunocytochemistry - Neurofibrillary tangles - Paired helical filaments - Tau protein
Supported by NIA AG06803, AG01136 and AG04145



Transglutaminase bonds in neurofibrillary tangles and paired helical filament tau early in Alzheimer's disease
http://www.sciencedirect.com/scidirimg/pdf_icon.gif

Steven M. Singera, Gina M. Zainellia, Maryam A. Norlunda, John M. Leea, b and Nancy A. MumaCorresponding Author Contact Information, E-mail The Corresponding Author, a
a The Department of Pharmacology, Loyola University Medical Center, 2160 S First Avenue, Maywood, IL 60153, USA
b The Department of Pathology, Loyola University Medical Center, Maywood, IL 60153, USA

Available online 1 December 2001.

Abstract

Transglutaminase-catalyzed var epsilon(γ-glutamyl)lysine cross-links exist in Alzheimer's disease (AD) paired helical filament (PHF) tau protein but not normal soluble tau. To test the hypothesis that these cross-links could play a role in the formation of neurofibrillary tangles (NFT), we used single- and double-label immunofluorescence confocal microscopy and immunoaffinity purification and immunoblotting to examine var epsilon(γ-glutamyl)lysine cross-links in AD and control brains. The number of neurons that are immunoreactive with an antibody directed at the var epsilon-(γ-glutamyl)lysine bond was significantly higher in AD cortex compared with age-matched controls and schizophrenics. PHF tau-directed antibodies AT8, MC-1 and PHF-1 co-localized with var epsilon(γ-glutamyl)lysine immunolabeling in AD NFT. Immunoaffinity purification and immunoblotting experiments demonstrated that PHF tau contains var epsilon(γ-glutamyl)lysine bonds in parietal and frontal cortex in AD. In control cases with NFT present in the entorhinal cortex and hippocampus, indicative of Braak and Braak stage II, var epsilon(γ-glutamyl)lysine bonds were present in PHF tau in parietal and frontal cortex, despite the lack of microscopically detectable NFT or senile plaques in these cortical regions. The presence of PHF tau with var epsilon(γ-glutamyl)lysine bonds in brain regions devoid of NFT in stage II (but regions, which would be expected to contain NFT in stage III) suggests that these bonds occur early in the formation of NFT.
Author Keywords: Alzheimer's disease; Cross-linking; Neurofibrillary tangle; Paired helical filament; Tauopathy; Transglutaminase
Abbreviations: AD, Alzheimer's disease; Mab, monoclonal antibody; NFT, neurofibrillary tangles; PHF, paired helical filaments

Article Outline


Am J Pathol. 1990 May; 136(5): 1069–1075.
PMCID: PMC1877428
Relative abundance of tau and neurofilament epitopes in hippocampal neurofibrillary tangles.
M. L. Schmidt, V. M. Lee, and J. Q. Trojanowski
Department of Pathology and Laboratory Medicine (Neuropathology), University of Pennsylvania School of Medicine, Philadelphia 19104-4283.
Neurofibrillary tangles (NFTs) derive, in part, from normal neuronal cytoskeletal proteins, ie, large portions of tau (tau) but only restricted segments of the peripheral domains of the high- and middle-molecular weight neurofilament subunits. To learn more about the events leading to the incorporation of tau and neurofilament epitopes into NFTs, the relative abundance of tau and NF determinants in these lesions was quantitatively analyzed in hippocampi from Alzheimer disease (AD) patients and age-matched controls using monoclonal antibodies specific for tau or for NF proteins. Immunostained NFTs appeared qualitatively the same in both AD and controls, ie, every epitope found in AD NFTs occurred also in the NFTs of the control patients. However, in hippocampi with only a few tangles, tau epitopes, but no NF epitopes, were detected in NFTs. In contrast, both tau and NF epitopes were present in those tangles that were found in hippocampi with abundant NFTs. Nevertheless, the number of tau-positive NFTs generally exceeded the number of NF-positive NFTs. These findings indicate that tau epitopes are more abundant than NF epitopes in NFTs and that the formation of NFTs may be linked to a derangement in the normal metabolism of tau that is more extensive than alterations in NF protein metabolism.
Volume 88, Number 6, 592-598, DOI: 10.1007/BF00296499

Unusual case of corticobasal degeneration with tau/Gallyas-positive neuronal and glial tangles

Abstract

A 74-year-old woman with corticobasal degeneration (CBD) had a 9-year history of progressive loss of strength and rigidity of her right hand and then arm, followed by speech difficulties, dyskinesia, rigidity, spasticity and weakness of the ipsilateral lower limb, ultimately also involving the apposite side. She later developed supranuclear gaze palsy. Her memory remained intact during most of the duration of her disease. Laboratory tests and anti-Parkinsonian medications were not helpful. At autopsy, frontal lobe atrophy, discoloration of putamen (Pt) and pallor of substantia nigra (Sn) were observed. Neuronal loss and gliosis were extensive in motor cortex and milder in frontal cortex, abruptly ending at the central sulcus and junction of cingulate gyrus. ldquoAchromaticrdquo neurons were present. Neuronal loss and gliosis were seen in Pt and Sn and corticobasal inclusions in Sn. Numerous Gallyas/tau-positive, Bielschowsky/ubiquitin-negative coil, sickle, or coma-shaped tangles and thread-like processes were found in affected cortex, Pt and Sn. Some of the tangles were in neurons, but most occurred in astroglia, and their processes. The presence of Gallyas/tau-positive glia in CBD may have the same diagnostic significance as in progressive supranuclear palsy, analogous to the argyrophilic ubiquinated inclusions in oligodendroglia in multisystem atrophy. We suggest that in CBD: (1) cytoskeletal protein metabolism in neurons and glia can simultaneously be perturbed in certain neurodegenerative diseases, and (2) the astrocytosis in CBD may not be simply a reactive process but an integral part of the disease.
Volume 77, Number 4, 430-436, DOI: 10.1007/BF00687379

Senile plaque neurites fail to demonstrate anti-paired helical filament and anti-microtubule-associated protein-tau immunoreactive proteins in the absence of neurofibrillary tangles in the neocortex

Abstract

Although much work has been directed recently towards unravelling the protein chemistry of neurofibrillary tangle (NFT) and senile plaque (SP) components in Alzheimer's disease, the pathogeneses of these lesions remains largely unknown and the problem of their relationship is unresolved. In particular, although paired helical filaments (PHF) have long been documented in SP neurites, we do not know if they are of pathogenetic relevance for the formation of the SP. To investigate the relationship between NFT and SP, we examined antigenic properties of proteins in SP neurites in neocortical tissues of patients with senile dementia of Alzheimer type, in the presence or absence of NFT in the same cortical area. We used two polyclonal antibodies directed against PHF and microtubule-associated protein (MAP)-tau and three monoclonal antibodies (MAbs) (RT97, BF10, 147) to phosphorylated epitopes of human neurofilament polypeptides, as well as the Gallyas silver impregnation method which specifically stains PHF in NFT and neurites. The main finding of our investigations consists in a differential pattern of immunoreactivity of SP neurites depending on the presence or absence of NFT in the neocortex. In the presence of NFT, there were numerous neuropil threads and SP neurites containing Gallyas-positive, as well as anti-PHF- and anti-tau-labelled material. In the absence of NFT in the neocortex there was a striking absence of any Gallyas-positive or PHF- and tau-immunoreactive structure in the cortical neuropil and in SP neurites, irrespective of the maturation stage of the SP. In contrast with these results, the number of neurites labelled by MAbs RT97, BF10 and 147 in SP and in the neuropil was apparently unaffected by the presence or absence of NFT. Amyloid in SP, remained consistently unstained by all antibodies of the panel as well as by the Gallyas stain. Our findings indicate that PHF and tau polypeptides are facultative components of SP neurites and suggest that the development of SP may occur independently of PHF pathology in neocortical neurons.
Key words  Senile plaque - Paired helical filaments - Microtubule-associated (MAP)-tau protein - Neurofibrillary tangles - Alzheimer
Supported by the Wellcome Trust and the Medical Research Council
Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease
Volume 1739, Issues 2-3, 3 January 2005, Pages 216-223
The Biology and Pathobiology of Tau

Review

Tau, tangles, and Alzheimer's disease
http://www.sciencedirect.com/scidirimg/pdf_icon.gif

Lester I. Bindera, b, Corresponding Author Contact Information, E-mail The Corresponding Author, Angela L. Guillozet-Bongaartsa, b, Francisco Garcia-Sierrac and Robert W. Berrya, b
aDepartment of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Avenue, Chicago, IL 60611, United States
bCognitive Neurology and Alzheimer's Disease Center, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Avenue, Chicago, IL 60611, United States
cDepartment of Cell Biology, Center of Research and Advanced Studies of the National Polytechnic Institute, Av. Instituto Politecnico Nacional 2508, CP 07360, Mexico City, Mexico
Received 30 August 2004;
accepted 31 August 2004.
Available online 15 September 2004.

Abstract

Neurofibrillary tangles (NFT) are comprised of the microtubule-associated protein tau, in the form of filamentous aggregates. In addition to the well-known changes in phosphorylation state, tau undergoes multiple truncations and shifts in conformation as it transforms from an unfolded monomer to the structured polymer characteristic of NFT. Truncations at both the amino- and carboxy-termini directly influence the conformation into which the molecule folds, and hence the ability of tau to polymerize into fibrils. Certain of these truncations may be due to cleavage by caspases as part of the apoptotic cascade. In this review, we discuss evidence that strongly suggests that these truncations occur in an orderly pattern and directly influence the ability of tau to polymerize into filaments.
Keywords: Neurofibrillary tangle; Tau; Alzheimer's disease

Article Outline

1.
2.
3.
4.
5.
6.
7.
8.
9.
Multiple isoforms of human microtubule-associated protein tau: sequences and localization in neurofibrillary tangles of Alzheimer's disease
http://www.sciencedirect.com/scidirimg/pdf_icon.gif

M. Goederta, M.G. Spillantinia, R. Jakesa, D. Rutherforda and R.A. Crowthera
a Medical Research Council Laboratory of Molecular Biology Hills Road Cambridge CB2 2QH England
Received 19 June 1989.
Available online 20 April 2004.

Abstract

We have determined the sequences of isoforms of human tau protein, which differ from previously reported forms by insertions of 29 or 58 amino acids in the amino-terminal region. Complementary DNA cloning shows that the insertions occur in combination with both three and four tandem repeats. RNAase protection assays indicate that transcripts encoding isoforms with the insertions are expressed in an adult-specific manner. Transcripts encoding four tandem repeats are also expressed in an adult-specific manner, whereas mRNAs encoding three tandem repeats are expressed throughout life, including in fetal brain. The levels of transcripts encoding the 29 or 58 amino acid inserts were not significantly changed in cerebral cortex from patients with Alzheimer's disease. Antisera raised against synthetic peptides corresponding to these different human tau isoforms demonstrate that multiple tau protein isoforms are incorporated into the neurofibrillary tangles of Alzheimer's disease.


http://www.sciencedirect.com/scidirimg/pdf_icon.gif


Neuron
Volume 3, Issue 4, October 1989, Pages 519-526

Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing mutant (P301L) tau protein http://www.med.upenn.edu/ins/gradprog/courses/insc_670_current_topics_in_neuropharmacology_05/1_Giass_Lewis_et_al.pdf

Science 24 August 2001: 
Vol. 293 no. 5534 pp. 1491-1495 
DOI: 10.1126/science.1062097
REPORT

Formation of Neurofibrillary Tangles in P301L Tau Transgenic Mice Induced by Aβ42 Fibrils

+Author Affiliations
1 Division of Psychiatry Research, University of Zürich, August Forel Strasse 1, 8008 Zürich, Switzerland.
2 Experimental Genetics Group, Center for Human Genetics, K. U. Leuven, Campus Gasthuisberg, Leuven, Belgium.

ABSTRACT

β-Amyloid plaques and neurofibrillary tangles (NFTs) are the defining neuropathological hallmarks of Alzheimer's disease, but their pathophysiological relation is unclear. Injection of β-amyloid Aβ42 fibrils into the brains of P301L mutant tau transgenic mice caused fivefold increases in the numbers of NFTs in cell bodies within the amygdala from where neurons project to the injection sites. Gallyas silver impregnation identified NFTs that contained tau phosphorylated at serine 212/threonine 214 and serine 422. NFTs were composed of twisted filaments and occurred in 6-month-old mice as early as 18 days after Aβ42injections. Our data support the hypothesis that Aβ42 fibrils can accelerate NFT formation in vivo.
* These authors contributed equally to this work.
 To whom correspondence should be addressed at the Division of Psychiatry Research, University of Zürich, August Forel Strasse 1, 8008 Zürich, Switzerland. E-mail: goetz@bli.unizh.ch
 To whom correspondence should be addressed at the Division of Psychiatry Research, University of Zürich, August Forel Strasse 1, 8008 Zürich, Switzerland. E-mail: nitsch@bli.unizh.ch
Nature Medicine  2, 783 - 787 (1996) 
doi:10.1038/nm0796-783

Alzheimer's disease hyperphosphorylated tau sequesters normal tau into tangles of filaments and disassembles microtubules

Alejandra del C. Alonso1, 2, 4, Inge Grundke-Iqbal1 & Khalid Iqbal1, 3
1New York State Institute for Basic Research in Developmental Disabilities,1050 Forest Hill Road, Staten Island, New York 10314 USA
2Departamento de Química Biológica, Facultad de Cs. Qs. - U.N.C., Ciudad Universitaria - Pab. Argentina, cc 61 - cp 5016, Cordoba, Argentina
3Correspondence should be addressed to K.I.
4A.d.C.A. present address: Departamento di Química Biólogica, Facultad de Cs. Qs. − U.N.C., Cuidad Universitaria − Pab. Argentina, cc 61 − cp 5016, Cordoba, Argentina
http://www.nature.com/images/spacer.gif
http://www.nature.com/nm/images/spacer_pink.gif
http://www.nature.com/images/spacer.gif
Microtubule−associated protein tau becomes abnormally hyperphosphorylated in Alzheimer's disease (AD) and accumulates as tangles of paired helical filaments in neurons undergoing degeneration. We now show that in solution normal tau associates with the AD hyperphosphorylated tau (AD P−tau) in a nonsaturable fashion, forming large tangles of filaments 3.3 plusminus 0.7 nm in diameter. These tangles, which are not detected in identically treated normal tau or AD P−tau alone, are made up of filaments several microns in length and are labeled with tau antibodies. Dephosphorylation with alkaline phosphatase abolishes the ability of AD P−tau to aggregate with normal tau and prevents tangle formation. AD P−tau disassembles microtubules assembled from normal tau and tubulin. These data provide insight into how the hyperphosphorylation of tau might lead to the formation of the neurofibrillary tangles and the degeneration of the affected neurons in AD.
http://www.nature.com/images/spacer.gif
http://www.nature.com/nm/images/spacer_pink.gif
http://www.nature.com/images/spacer.gif

REFERENCES
Tomlinson, B.E., Blessed, G. & Roth, M. Observations on the brains of demented old people. J. Neurol. Sci. 11, 205−242 (1970). | Article | PubMed  | ISI | ChemPort |
Alafuzoff, I., Iqbal, K., Friden, H., Adolfsson, R. & Winblad, B. Histopathological criteria for progressive dementia disorders: Clinical-pathological correlation and classification by multivariate data analysis.Acta Neuropathol. 74, 209−225 (1987). | PubMed  | ISI | ChemPort |
Grundke-Iqbal, I. et al. Abnormal phosphorylation of the microtubule-associated protein tau in Alzheimer cytoskeletal pathology. Proc. Natl. Acad. Sci. USA 83, 4913−4917 (1986). | PubMed  | ChemPort |
Grundke-Iqbal, I. et al. Microtubule-associated protein tau: A component of Alzheimer paired helical filaments. J. Biol. Chem. 261, 6084−6089 (1986). | PubMed  | ChemPort |
Köpke, E. et al. Microtubule associated protein tau: Abnormal phosphorylation of non-paired helical filament pool in Alzheimer disease.J. Biol. Chem. 268, 24374−24384 (1993). | PubMed  | ISI |
Khatoon, S., Grundke-Iqbal, I. & Iqbal, K. Brain levels of microtubule associated protein tau are elevated in Alzheimer's disease brain: A radioimmunoslot-blot assay for nanograms of the protein. J. Neurochem.59, 750−753 (1992). | PubMed  | ISI | ChemPort |
Alonso, A.d.C., Zaidi, T., Grundke-Iqbal, I. & Iqbal, K. Role of abnormally phosphorylated tau in the breakdown of microtubules in Alzheimer disease. Proc. Natl. Acad. Sci. USA 91, 5562−5566 (1994). | PubMed  | ChemPort |
Iqbal, K., Zaidi, T., Bancher, C. & Grundke-Iqbal, I. Alzheimer paired helical filaments: Restoration of the biological activity by dephosphorylation. FEBS Lett. 349, 104−108 (1994). | Article | PubMed | ISI | ChemPort |
Wang, J.-Z., Gong, C.-X., Zaidi, T., Grundke-Iqbal, I. & Iqbal, K. Dephosphorylation of Alzheimer paired helical filaments by protein phosphatase-2A and-2B. J. Biol. Chem. 270, 4854−4860 (1995). | Article | PubMed  | ISI | ChemPort |
Iqbal, K. et al. Defective brain microtubule assembly in Alzheimer's disease. Lancet 2, 421−426 (1986). | Article | PubMed | ISI | ChemPort |
Ruben, G.C. et al. The microtubule associated protein tau forms a triple stranded left-hand helical polymer. J. Biol. Chem. 266, 22019−22027 (1991). | PubMed  | ISI | ChemPort |
Wang, J.-Z., Grundke-Iqbal, I. & Iqbal, K. Glycosylation of microtubule associated protein tau: An abnormal posttranslational modification in Alzheimer disease. Nature Medicine, see accompanying manuscript.
Ruben, G.C., Iqbal, K., Wisniewski, H.M., Johnson, J.E., Jr. & Grundke-Iqbal, I. Alzheimer neurofibrillary tangles contain 2.1-nm filaments structurally identical to the microtubule associated protein tau: A high resolution transmission electron microscope study of tangles and senile plaque core amyloid. Brain Res. 590, 164−179 (1992). | Article | ISI | ChemPort |
Wille, H., Drewes, G., Biernat, J., Mandelkow, E.M. & Mandelkow, E. Alzheimer-like paired helical filaments and antiparallel dimers formed from microtubule-associated protein tau in vitro. J. Cell Biol. 118, 573−584 (1992). | Article | PubMed  | ISI | ChemPort |
Crowther, R.A., Olesen, O.F., Jakes, R. & Goedert, M. The microtubule binding repeats of tau protein assemble into filaments like those found in Alzheimer's disease. FEBS Lett. 309, 199−202 (1992). | Article | PubMed  | ISI | ChemPort |
Crowther, R.A., Olesen, O.F., Smith, M., Jakes, R. & Goedert, M. Assembly of Alzheimer-like filaments from full-length tau protein. FEBS Lett. 337, 135−138 (1994). | Article | PubMed  | ISI | ChemPort |
Schweers, O., Mandelkow, E.M., Biernat, J. & Mandelkow, E. Oxidation of cysteine-322 in the repeat domain of microtubule-associated protein taucontrols the in vitro assembly of paired helical filaments. Proc. Natl. Acad. Sci. USA 92, 8463−8467 (1995). | PubMed  | ChemPort |
Bancher, C. et al. Accumulation of abnormally phosphorylated tau precedes the formation of neurofibrillary tangles in Alzheimer's disease.Brain Res. 477, 90−99 (1989). | Article | PubMed  | ISI | ChemPort |
Gong, C.-X., Singh, T.J., Grundke-Iqbal, I. & Iqbal, K. Phosphoprotein phosphatase activities in Alzheimer disease brain. J. Neurochem. 61, 921−927 (1993). | PubMed  | ISI | ChemPort |
Gong, C.-X. et al. Phosphatase activity towards abnormally phosphorylated tau: Decrease in Alzheimer disease brain. J. Neurochem. 65, 732−738,(1995). | PubMed  | ISI | ChemPort |
Ruben, G.C., Harris, E.D.Jr. & Nagase, H. Electron microscope studies of free and proteinase-bound duck ovostatins (ovomacroglobins): Model of ovostatin structure and its transformation upon proteolysis. J. Biol. Chem.263, 2861−2869 (1988). | PubMed  | ISI | ChemPort |
Wrzolek, M.A. et al. Immune electron microscopic characterization of monoclonal antibodies to Alzheimer neurofibrillary tangles. Am. J. Pathol.141, 343−355 (1992). | PubMed  | ISI | ChemPort |
Grundke-Iqbal, I. et al. Microtubule-associated polypeptides tau are altered in Alzheimer paired helical filaments. Mol. Brain Res. 4, 43−52 (1988). | Article | ChemPort |
Alonso, A. del C., Grundke-Iqbal, I. & Iqbal, K. Bovine and human tau, highly homologous but less crossreactive: Implications for Alzheimer disease. Brain Res. 31, 194−200 (1995). | Article | ChemPort |
Bensadoun, A. & Weinstein, D. Assay of proteins in the presence of interfering materials. Anal. Biochem. 70, 241−250 (1976). | PubMed | ISI | ChemPort |
Shelanski, M.L., Gaskin, F. & Cantor, C.R. Microtubule assembly in the absence of added nucleotides. Proc. Natl. Acad. Sci. USA 70, 765−768 (1973). | PubMed  | ChemPort |
Sloboda, R.D. & Rosenbaum, J.L. Decoration and stabilization of intact, smooth-walled microtubules with microtubule-associated proteins.Biochemistry 18, 48−55 (1979). | PubMed  | ISI | ChemPort |
Biernat, J. et al. The switch of tau protein to an Alzheimer-like state includes the phosphorylation of two serine-proline motifs upstream of the microtubule binding region. EMBO J. 11, 1593−1597 (1992). | PubMed  | ISI | ChemPort |
Greenberg, S.G., Davies, P., Schein, J.D. & Binder, L. Hydrofluoric acid-treated tau. PHF proteins display the same biochemical properties as normal tau. J. Biol. Chem. 267, 564−569 (1992). | PubMed | ISI | ChemPort |
Otvos, L. et al. Monoclonal antibody PHF-1 recognizes tau protein phosphorylated at serine residue 396 and residue 404. J. Neurosci. Res.39, 669−673 (1994). | PubMed  | ISI | ChemPort |
Accumulation of abnormally phosphorylated τ precedes the formation of neurofibrillary tangles in Alzheimer's disease
http://www.sciencedirect.com/scidirimg/pdf_icon.gif

C. Banchera, Corresponding Author Contact Information, C. Brunnera, H. Lassmanna, H. Budkaa, K. Jellingerc, G. Wicheb, F. Seitelbergera, I. Grundke-Iqbald, K. Iqbald and H. M. Wisniewskid
a Neurological Institute, University of Vienna, Vienna, Austria
b Institute for Biochemistry, University of Vienna, Vienna, Austria
c Ludwig Boltzmann Institute for Clinical Neurobiology, Lainz Geriatric Hospital, Vienna, Austria
d New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, U.S.A.
Accepted 28 June 1988. ;
Available online 14 March 2003.

Abstract

The intraneuronal accumulation of paired helical filaments in the form of neurofibrillary tangles is one hallmark of the brain pathology in Alzheimer's disease. At certain predilection sites, a small number of similar lesions are also present in the brains of the majority of aged non-demented individuals. As suggested by several studies before, these abnormal cytoskeletal structures contain determinants of microtubule-associated protein τ and ubiquitin. The present study uses a morphological classification of neurofibrillary tangles into different stages of maturation, as suggested by Alzheimer in 1911, and shows by quantitative immunocytochemistry that early stages of neurofibrillary degeneration contain abnormally phosphorylated τ. Immunoreactivity for the altered τ is seen not only in tangles but also in the cytoplasm of some nerve cells lacking neurofibrillary tangles. Similar numbers of such immunoreactive neurons without tangles are present in age-matched non-demented individuals as in Alzheimer cases, but are absent in young controls. In contrast, incorporation of an epitope, recognized by a monoclonal antibody (3–39) raised to paired helical filaments, which is directed against a determinant residing in the 50–65 amino acid residue region of ubiquitin occurs late in the process of tangle maturation and is most pronounced in extracellular ‘ghost tangles’. It is suggested that the accumulation of abnormally phosphorylated τ is one of the earliest cytoskeletal changes in the process of tangle formation. Exposure of certain ubiquitin epitopes in the pathological fibers may reflect an unsuccessful attempt of proteolytic degradation.
Author Keywords: Alzheimer's disease; Neurofibrillary tangle; Paired helical filament; Protein phosphorylation; Tau; Ubiquitin

Neurofibrillary tangles of Alzheimer disease share antigenic determinants with the axonal microtubule-associated protein tau (tau)

Abstract

The relationship of the neurofibrillary tangle, found in Alzheimer disease and aged brains, to normal or abnormal cytoskeletal proteins remains elusive. Although immunohistochemical studies have yielded disparate results, most antigenic determinants localized to neurofibrillary tangles are cytoskeletal constituents normally present in neuronal perikarya or dendrites. We report light and electron microscopic immunolabeling of neurofibrillary tangles by a monoclonal antibody to the microtubule-associated protein tau (tau). Dephosphorylation of tissue slices not only increased the number of tau-positive tangles but also produced marked positive immunoreactivity of neuritic plaques. The localization of tau, an axonal protein, to neurofibrillary tangles in the perikaryon in particular suggests that abnormal synthesis, modification, or aggregation of tau may induce aberrant cytoskeletal--cell organelle interactions, subsequent interference with axonal flow, and resultant tangle formation.

Caspase cleavage of tau: Linking amyloid and neurofibrillary tangles in Alzheimer's disease

+Author Affiliations
Departments of *Cell and Molecular Biology and §Biochemistry and Molecular Pharmacology; Cell Death Regulation Laboratory, Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine; and Cognitive Neurology and Alzheimer's Disease Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
Edited by L. L. Iversen, University of Oxford, Oxford, United Kingdom, and approved June 16, 2003 (received for review January 23, 2003)

Abstract

The principal pathological features of Alzheimer's disease (AD) are extracellular amyloid plaques and intracellular neurofibrillary tangles, the latter composed of the microtubule-binding protein tau assembled into paired helical and straight filaments. Recent studies suggest that these pathological entities may be functionally linked, although the mechanisms by which amyloid deposition promotes pathological tau filament assembly are poorly understood. Here, we report that tau is proteolyzed by multiple caspases at a highly conserved aspartate residue (Asp421) in its C terminus in vitro and in neurons treated with amyloid-β (Aβ) (1–42) peptide. Tau is rapidly cleaved at Asp421 in Aβ-treated neurons (within 2 h), and its proteolysis appears to precede the nuclear events of apoptosis. We also demonstrate that caspase cleavage of tau generates a truncated protein that lacks its C-terminal 20 amino acids and assembles more rapidly and more extensively into tau filaments in vitro than wild-type tau. Using a monoclonal antibody that specifically recognizes tau truncated at Asp421, we show that tau is proteolytically cleaved at this site in the fibrillar pathologies of AD brain. Taken together, our results suggest a novel mechanism linking amyloid deposition and neurofibrillary tangles in AD: Aβ peptides promote pathological tau filament assembly in neurons by triggering caspase cleavage of tau and generating a proteolytic product with enhanced polymerization kinetics.

Footnotes

** To whom correspondence should be addressed at: Division of Endocrinology, Tarry 15-755, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611. E-mail: v-cryns@northwestern.edu.
 T.C.G. and F.C. contributed equally to this work.
 L.I.B. and V.L.C. contributed equally to this work.
This paper was submitted directly (Track II) to the PNAS office.
Abbreviations: AD, Alzheimer's disease; NFTs, neurofibrillary tangles; Aβ, amyloid-β.
Alz-50, Ubiquitin and Tau Immunoreactivity of Neurofibrillary Tangles, Pick Bodies and Lewy Bodies
Love, S M.B.B.Ch., Ph.D.; Saitoh, T Ph.D.; Quijada, S; Cole, G M Ph.D.; Terry, R D M.D.
Collapse Box
Abstract
Immunocytochemical and quantitative immunochemical techniques were used to study the expression of Alz-50 antigen, ubiquitin and Tau in neurologic disorders characterized by the formation of filamentous neuronal inclusions. Alz-50, anti-ubiquitin and Tau-1 immunostained the intraneuronal neurofibrillary tangles and the neuritic component of plaques, both in Alzheimer's disease and in the brains of patients without dementia, but extraneuronal tangles were largely unstained. These antibodies also reacted with Pick bodies, and with the neurofibrillary tangles of Kufs' disease and Guam Parkinsonism-dementia. In sections from the brain of a patient with progressive supranuclear palsy, virtually all of the tangles were immunostained with Tau-1 but only a few with Alz-50 or anti-ubiquitin. Anti-ubiquitin also labelled Lewy bodies and the inclusions of granulovacuolar degeneration. Quantitative analysis of immunoblots of homogenized frontal cortex showed significantly more Alz-50 antigen in the brains of patients with Alzheimer's and Pick's disease than in controls. The level of this antigen was increased both in the crude homogenates and in the cytosolic fraction. Ubiquitin immunoreactivity was increased only in the brains of patients with Alzheimer's disease and then only in the crude homogenates. The finding that antigenic determinants for Alz-50, anti-ubiquitin and Tau-1 are shared by several filamentous neuronal inclusions occurring in diverse neurologic disorders may reflect common metabolic defects underlying the formation of these inclusions, or common metabolic responses to their presence.
(C) 1988 American Association of Neuropathologists, Inc
EMBO J. 1990 November; 9(11): 3539–3544.
PMCID: PMC552103
Phosphorylation of microtubule-associated protein tau: identification of the site for Ca2(+)-calmodulin dependent kinase and relationship with tau phosphorylation in Alzheimer tangles.
B Steiner, E M Mandelkow, J Biernat, N Gustke, H E Meyer, B Schmidt, G Mieskes, H D Söling, D Drechsel, M W Kirschner
, et al.
Max-Planck-Unit for Structural Molecular Biology, Hamburg, FRG.
The microtubule array in neuronal cells undergoes extensive growth, dynamics and rearrangements during neurite outgrowth. While little is known about how these changes are regulated, microtubule-associated proteins (MAPs) including tau protein are likely to perform an important role. Tau is one of the MAPs in mammalian brain. When isolated it is usually a mixture of several isoforms containing between 341 and 441 residues that arise from alternative splicing. Tau can be phosphorylated by several protein kinases. Phosphorylation at certain sites results in major structural and functional changes, as seen by changes in electrophoretic mobility, interaction with microtubules, molecular length and elasticity. Here we show that the sites of phosphorylation by four kinases (PKA, PKC, CK and CaMK) all lie in the C-terminal microtubule-binding half of tau, but only the phosphorylation by CaM kinase shows the pronounced shift in electrophoretic mobility characteristic for tau from Alzheimer neurofibrillary tangles. By using a combination of limited proteolysis, protein sequencing and protein engineering we show that a single phosphorylation site is responsible for this shift, located at Ser 405 in the C-terminal tail of the protein outside the region of internal repeats. Phosphorylation at this site not only reduces the electrophoretic mobility of tau, it also makes the protein long and stiff, as shown earlier. The site is likely to be phosphorylated in tau from Alzheimer neurofibrillary tangles.

Recognition of tau epitopes by anti-neurofilament antibodies that bind to Alzheimer neurofibrillary tangles

Abstract

Eleven anti-neurofilament (anti-NF) monoclonal antibodies were studied for their reactivity with heat-stable, microtubule-associated proteins and Alzheimer neurofibrillary tangles (ANT). On immunoblots of NF proteins, the antibodies recognized epitopes that were variably sensitive to Escherichia coli alkaline phosphatase. Eight of the antibodies showed reactivity with ANT and decreased binding to electroblotted NF after phosphatase treatment. The same eight antibodies reacted with tau proteins from bovine and rat brain, binding to tau proteins was also substantially reduced by phosphatase. Of the eight antibodies that bound to animal tau proteins, five also bound to tau proteins from normal human brain. All of the antibodies that bound to animal tau proteins stained ANT in frozen tissue sections. Brief treatment of tissue sections with trypsin in most cases enhanced antibody binding to ANT. All antibodies that lacked reactivity with tau proteins failed to bind ANT. Phosphatase treatment of Alzheimer tissue sections did not change the immunoreactivity of ANT and neurites in senile plaques with ANT-reactive, anti-NF antibodies, except for two antibodies that showed decreased binding to ANT. In contrast, axonal staining was decreased or eliminated by phosphatase treatment, similar to the response of electroblotted NF and tau proteins. These results suggest that staining of ANT by anti-NF antibodies may be due to cross-reaction of anti-NF with epitopes in tau proteins, the epitopes in axons, NF, and tau are sensitive to the effect of phosphatase, whereas the majority of those in ANT are not, and some of the epitopes in ANT that are shared with NF and tau proteins are not readily accessible to antibody binding.

Abstract

The history of Alzheimer’s disease (AD) presents a difficulty in a book that is devoted to the cholinergic system. Indeed, this history emphasizes the tale of plaques and bundles; the heroes of the tale are Alois Alzheimer and Gaetano Perusini, and their discoveries concern the amyloid “cement”; this tale, told in section 1 of this introduction, does not seem to relate to cholinergicity. Similarly, as a whole, the chapter focuses on neurocellular degeneration and degenerative proteins as the basis of AD rather than on cholinergicity. Yet, there is a strong link between AD and the cholinergic system in view of the apparent preference of plaques, bundles, and degenerative proteins for the central cholinergic neurons. This matter and its consequences are discussed in section 2 of this introduction and expanded upon in section K (Wong et al., 2006).

Vascular variant of prion protein cerebral amyloidosis with tau-positive neurofibrillary tangles: the phenotype of the stop codon 145 mutation in PRNP

+Author Affiliations
Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis 46202-5120, USA.

Abstract

Deposition of PrP amyloid in cerebral vessels in conjunction with neurofibrillary lesions is the neuropathologic hallmark of the dementia associated with a stop mutation at codon 145 of PRNP, the gene encoding the prion protein (PrP). In this disorder, the vascular amyloid in tissue sections and the approximately 7.5-kDa fragment extracted from amyloid are labeled by antibodies to epitopes located in the PrP sequence including amino acids 90-147. Amyloid-laden vessels are also labeled by antibodies against the C terminus, suggesting that PrP from the normal allele is involved in the pathologic process. Abundant neurofibrillary lesions are present in the cerebral gray matter. They are composed of paired helical filaments, are labeled with antibodies that recognize multiple phosphorylation sites in tau protein, and are similar to those observed in Alzheimer disease. A PrP cerebral amyloid angiopathy has not been reported in diseases caused by PRNP mutations or in human transmissible spongiform encephalopathies; we propose to name this phenotype PrP cerebral amyloid angiopathy (PrP-CAA).
The level of cerebrospinal fluid tau correlates with neurofibrillary tangles in Alzheimer's disease
Tapiola, Tero1; Overmyer, Margit1; Lehtovirta, Maarit1; Helisalmi, Seppo1,3; Ramberg, Jarmo4; Alafuzoff, Irina1; Riekkinen, Paavo Sr1,2; Soininen, Hilkka1,5

Abstract
WE measured tau concentrations in cerebrospinal fluid (CSF) samples taken during the lifetime of 43 patients with Alzheimer's disease (AD) and correlated these values with neurofibrillary tangle (NFT) scores as well as glial fibrillary acidic protein (GFAP) expression as a marker of astrocytosis in the brain post-mortem. The CSF tau values showed a positive correlation with neocortical NFT scores (r = 0.44, p < 0.005), while GFAP immmunoreactivity did not correlate with CSF tau. This study reveals a high variation in CSF tau values in patients with neuropathologically confirmed AD (range 194-1539 pg/ml) and indicates that high CSF tau values in the late phase of Alzheimer's disease predict severe neurodegeneration as evidenced by increased NFT scores.
(The FASEB Journal. 2005;19:869-871.)
© 2005 FASEB

Proteomic analysis of neurofibrillary tangles in Alzheimer disease identifies GAPDH as a detergent-insoluble paired helical filament tau binding protein

Qin Wang, Randall L. Woltjer, P. J. Cimino, Catherine Pan, Kathleen S. Montine, Jing Zhang and Thomas J. Montine1
Department of Pathology, Division of Neuropathology, University of Washington, Seattle, Washington, USA
1 Correspondence: Department of Pathology, Division of Neuropathology, University of Washington, Seattle, Washington 98105, USA. E-mail: tmontine@u.washington.edu
SPECIFIC AIMS
Neurofibrillary tangles (NFTs), hallmark pathologic inclusions of Alzheimer’s disease (AD) most closely associated with dementia, are composed primarily of an aberrant form of the microtubule binding protein tau. The aim of this study was to perform proteomic analysis using liquid chromatography (LC)/mass spectrometry (MS)-MS of tryptic digests of laser-captured NFTs to identify other NFT-associated proteins and to pursue immunohistochemical and biochemical validation and characterization of selected proteins identified by this unbiased survey.
Pathological Tau Tangles Localize to Focal Cortical Dysplasia in Older Patients
Arjune Sen1,
Maria Thom1,2,
Lillian Martinian1,2,
Brian Harding3,
J. Helen Cross4,
Margareta Nikolic5,
Sanjay M. Sisodiya1
DOI: 10.1111/j.1528-1167.2007.01107.x
Keywords:
cdk5;
Epilepsy;
Focal cortical dysplasia;
GSK3β;
Neurodegeneration;
Tau
Summary:Purpose: Reactivation of neurodevelopmental processes may contribute to neurodegeneration. For example, the proteins cyclin dependent kinase 5 (cdk5) and glycogen synthase kinase 3 beta (GSK3β), which are essential to normal cortical development, can hyperphosphorylate tau and might contribute to the pathogenesis of Alzheimer's disease. Focal cortical dysplasia (FCD) is an important neurodevelopmental cause of refractory human epilepsy within which dysplastic neurons exhibit increased immunoreactivity for cdk5 and GSK3β as well as neurofilamentous accumulations. We therefore hypothesized that the developmentally abnormal cortex of FCD might be more susceptible to tau-mediated neurodegeneration than adjacent histologically normal cortex.
Materials and Methods: We examined a series of 15 cases of FCD, spanning a wide age range, for β-amyloid, pathologically phosphorylated tau and neurofibrillary tangles using silver staining, immunohistochemistry for tau, AT8, RD3, RD4 and two-dimensional cell counting.
Results:β-amyloid plaques, aberrantly phosphorylated tau and neurofibrillary tangles are only found in older patients. The hyperphosphorylated tau tangles are confined to dysplastic neurons. Immunoreactivity for 3- and 4-repeat tau was again only detected within regions of FCD in older patients. With increasing age, the dysplastic cortex became hypocellular and a higher proportion of dysplastic neurons exhibited pathological tau phosphorylation.
Conclusions: In older patients, FCD appears more susceptible to formation of pathologically phosphorylated tau neurofibrillary tangles than adjacent histologically normal cortex. Our results suggest a novel convergence of pathological neurodevelopment with pathological age-related neurodegeneration.

No comments:

Post a Comment