Tau protein

Last updated
MAPT
Available structures
PDB Ortholog search: PDBe RCSB
Identifiers
Aliases MAPT , DDPAC, FTDP-17, MAPTL, MSTD, MTBT1, MTBT2, PPND, PPP1R103, TAU, microtubule associated protein tau, Tau proteins, tau-40
External IDs OMIM: 157140 MGI: 97180 HomoloGene: 74962 GeneCards: MAPT
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_001038609
NM_010838
NM_001285454
NM_001285455
NM_001285456

RefSeq (protein)
Location (UCSC) Chr 17: 45.89 – 46.03 Mb Chr 11: 104.23 – 104.33 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

The tau proteins (abbreviated from tubulin associated unit [5] ) are a group of six highly soluble protein isoforms produced by alternative splicing from the gene MAPT (microtubule-associated protein tau). [6] [7] They have roles primarily in maintaining the stability of microtubules in axons and are abundant in the neurons of the central nervous system (CNS), where the cerebral cortex has the highest abundance. [8] They are less common elsewhere but are also expressed at very low levels in CNS astrocytes and oligodendrocytes. [9]

Pathologies and dementias of the nervous system such as Alzheimer's disease and Parkinson's disease [10] are associated with tau proteins that have become hyperphosphorylated insoluble aggregates called neurofibrillary tangles. The tau proteins were identified in 1975 as heat-stable proteins essential for microtubule assembly, [5] [11] and since then they have been characterized as intrinsically disordered proteins. [12]

Neurons were grown in tissue culture and stained with antibody to MAP2 protein in green and MAP tau in red using the immunofluorescence technique. MAP2 is found only in dendrites and perikarya, while tau is found not only in the dendrites and perikarya but also in axons. As a result, axons appear red while the dendrites and perikarya appear yellow, due to superimposition of the red and green signals. DNA is shown in blue using the DAPI stain which highlights the nuclei. Image courtesy EnCor Biotechnology Inc. MAP2-tau in neurons.jpg
Neurons were grown in tissue culture and stained with antibody to MAP2 protein in green and MAP tau in red using the immunofluorescence technique. MAP2 is found only in dendrites and perikarya, while tau is found not only in the dendrites and perikarya but also in axons. As a result, axons appear red while the dendrites and perikarya appear yellow, due to superimposition of the red and green signals. DNA is shown in blue using the DAPI stain which highlights the nuclei. Image courtesy EnCor Biotechnology Inc.

Function

Microtubule stabilization

Tau proteins are found more often in neurons than in non-neuronal cells in humans. One of tau's main functions is to modulate the stability of axonal microtubules. [11] [13] Other nervous system microtubule-associated proteins (MAPs) may perform similar functions, as suggested by tau knockout mice that did not show abnormalities in brain development – possibly because of compensation in tau deficiency by other MAPs. [14] [15] [16]

Although tau is present in dendrites at low levels, where it is involved in postsynaptic scaffolding, [17] it is active primarily in the distal portions of axons, where it provides microtubule stabilization but also flexibility as needed. Tau proteins interact with tubulin to stabilize microtubules and promote tubulin assembly into microtubules. [11] Tau has two ways of controlling microtubule stability: isoforms and phosphorylation.

In addition to its microtubule-stabilizing function, Tau has also been found to recruit signaling proteins and to regulate microtubule-mediated axonal transport. [18]

mRNA translation

Tau is a negative regulator of mRNA translation in Drosophila [13] , mouse, [19] and human [20] brains, through its binding to ribosomes, which results in impaired ribosomal function, [21] reduction of protein synthesis and altered synaptic function. [13] [20] Tau interacts specifically with several ribosomal proteins, including the crucial regulator of translation rpS6. [22]

Behavior

The primary non-cellular functions of tau is to negatively regulate long-term memory [13] and to facilitate habituation (a form of non-associative learning), [13] two higher and more integrated physiological functions. Since regulation of tau is critical for memory, this could explain the linkage between tauopathies and cognitive impairment.

In mice, while the reported tau knockout strains present without overt phenotype when young, [14] [23] [24] when aged, they show some muscle weakness, hyperactivity, and impaired fear conditioning. [25] However, neither spatial learning in mice, [25] [26] [27] nor short-term memory (learning) in Drosophila [13] seems to be affected by the absence of tau.

In addition, tau knockout mice have abnormal sleep-wake cycle, with increased wakefulness periods and decreased non-rapid eye movements (NREM) sleep time. [28]

Other functions

Other typical functions of tau include cellular signalling, neuronal development, neuroprotection and apoptosis. [15] Atypical, non-standard roles of tau [29] are also under current investigation, such as its involvement in chromosome stability, its interaction with the cellular transcriptome, its interaction with other cytoskeletal or synaptic proteins, its involvement in myelination or in brain insulin signaling, its role in the exposure to chronic stress and in depression, etc.

Genetics

In humans, the MAPT gene for encoding tau protein is located on chromosome 17q21, containing 16 exons. [30] The major tau protein in the human brain is encoded by 11 exons. Exons 2, 3 and 10 are alternatively spliced, which leads to the formation of six tau isoforms. [31] In the human brain, tau proteins constitute a family of six isoforms with a range of 352–441 amino acids. Tau isoforms are different in having either zero, one, or two inserts of 29 amino acids at the N-terminal part (exons 2 and 3) and three or four repeat-regions at the C-terminal part (exon 10). Thus, the longest isoform in the CNS has four repeats (R1, R2, R3 and R4) and two inserts (441 amino acids total), while the shortest isoform has three repeats (R1, R3 and R4) and no insert (352 amino acids total).

The MAPT gene has two haplogroups, H1 and H2, in which the gene appears in inverted orientations. Haplogroup H2 is common only in Europe and in people with European ancestry. Haplogroup H1 appears to be associated with increased probability of certain dementias, such as Alzheimer's disease. The presence of both haplogroups in Europe means that recombination between inverted haplotypes can result in the lack of one of the functioning copies of the gene, resulting in congenital defects. [32] [33] [34] [35]

Structure

Six tau isoforms exist in human brain tissue, and they are distinguished by their number of binding domains. Three isoforms have three binding domains and the other three have four binding domains. The binding domains are located in the carboxy-terminus of the protein and are positively charged (allowing it to bind to the negatively charged microtubule). The isoforms with four binding domains are better at stabilizing microtubules than those with three binding domains. Tau is a phosphoprotein with 79 potential serine (Ser) and threonine (Thr) phosphorylation sites on the longest tau isoform. Phosphorylation has been reported on approximately 30 of these sites in normal tau proteins. [36]

Phosphorylation of tau is regulated by a host of kinases, including PKN, a serine/threonine kinase. When PKN is activated, it phosphorylates tau, resulting in disruption of microtubule organization. [37] Phosphorylation of tau is also developmentally regulated. For example, fetal tau is more highly phosphorylated in the embryonic CNS than adult tau. [38] The degree of phosphorylation in all six isoforms decreases with age due to the activation of phosphatases. [39] Like kinases, phosphatases too play a role in regulating the phosphorylation of tau. For example, PP2A and PP2B are both present in human brain tissue and have the ability to dephosphorylate Ser396. [40] The binding of these phosphatases to tau affects tau's association with microtubules.

Phosphorylation of tau has also been suggested to be regulated by O-GlcNAc modification at various Ser and Thr residues. [41]

Mechanism

The accumulation of hyperphosphorylated tau in neurons is associated with neurofibrillary degeneration. [42] The actual mechanism of how tau propagates from one cell to another is not well identified. Also, other mechanisms, including tau release and toxicity, are unclear. As tau aggregates, it replaces tubulin, which in turn enhances fibrilization of tau. [43] Several propagation methods have been proposed that occur by synaptic contact such as synaptic cell adhesion proteins, neuronal activity and other synaptic and non-synaptic mechanisms. [44] The mechanism of tau aggregation is still not completely elucidated, but several factors favor this process, including tau phosphorylation and zinc ions. [45] [46]

Release

Tau involves in uptake and release process, which is known as seeding. Uptake of tau protein mechanism requires the presence of heparan sulfate proteoglycans at the cell surface, which happen by macropinocytosis. [47] On the other hand, tau release depends on neuronal activity. Many factors influence tau release, for example, type of isoforms or MAPT mutations that change the extracellular level of tau. [48] According to Asai and his colleagues, the spreading of tau protein occurs from the entorhinal cortex to the hippocampal region in the early stages of the disease. They also suggested that microglia were also involved in the transport process, and their actual role is still unknown. [49]

Toxicity

Tau causes toxic effects through its accumulation inside cells. Many enzymes are involved in toxicity mechanism such as PAR-1 kinase. This enzyme stimulates phosphorylation of serine 262 and 356, which in turn leads to activate other kinases (GSK-3 and CDK5) that cause disease-associated phosphoepitopes. [50] The degree of toxicity is affected by different factors, such as the degree of microtubule binding. [51] [52] Toxicity could also happen by neurofibrillary tangles (NFTs), which leads to cell death and cognitive decline.

Clinical significance

Hyperphosphorylation of the tau protein (tau inclusions, pTau) can result in the self-assembly of tangles of paired helical filaments and straight filaments, which are involved in the pathogenesis of Alzheimer's disease, frontotemporal dementia and other tauopathies. [53] All of the six tau isoforms are present in an often hyperphosphorylated state in paired helical filaments in the Alzheimer's disease brain. In other neurodegenerative diseases, the deposition of aggregates enriched in certain tau isoforms has been reported. When misfolded, this otherwise very soluble protein can form extremely insoluble aggregates that contribute to a number of neurodegenerative diseases. Tau protein has a direct effect on the breakdown of a living cell caused by tangles that form and block nerve synapses. [54]

Gender-specific tau gene expression across different regions of the human brain has recently been implicated in gender differences in the manifestations and risk for tauopathies. [55] Some aspects of how the disease functions also suggest that it has some similarities to prion proteins. [56]

Tau hypothesis of Alzheimer's disease

The tau hypothesis states that excessive or abnormal phosphorylation of tau results in the transformation of normal adult tau into paired-helical-filament (PHF) tau and neurofibrillary tangles (NFTs). [57] The stage of the disease determines NFTs' phosphorylation. In AD, at least 19 amino acids are phosphorylated; pre-NFT phosphorylation occurs at serine 199, 202 and 409, while intra-NFT phosphorylation happens at serine 396 and threonine 231. [58] Through its isoforms and phosphorylation, tau protein interacts with tubulin to stabilize microtubule assembly. All of the six tau isoforms are present in an often hyperphosphorylated state in paired helical filaments (PHFs) in the AD brain.

Tau mutations have many consequences, including microtubule dysfunction and alteration of the expression level of tau isoforms. [59] Mutations that alter function and isoform expression of tau lead to hyperphosphorylation. The process of tau aggregation in the absence of mutations is not known but might result from increased phosphorylation, protease action or exposure to polyanions, such as glycosaminoglycans. Hyperphosphorylated tau disassembles microtubules and sequesters normal tau, MAPT 1 (microtubule associated protein tau 1), MAPT 2 and ubiquitin into tangles of PHFs. This insoluble structure damages cytoplasmic functions and interferes with axonal transport, which can lead to cell death. [60] [54]

Hyperphosphorylated forms of tau protein are the main component of PHFs of NFTs in the brain of AD patients. It has been well demonstrated that regions of tau six-residue segments, namely PHF6 (VQIVYK) and PHF6* (VQIINK), can form tau PHF aggregation in AD. Apart from the PHF6, some other residue sites like Ser285, Ser289, Ser293, Ser305 and Tyr310, located near the C-terminal of the PHF6 sequences, play key roles in the phosphorylation of tau. [61] Hyperphosphorylated tau differs in its sensitivity and its kinase as well as alkaline phosphatase activity [62] and is, along with beta-amyloid, a component of the pathologic lesion seen in Alzheimer disease. [63] [64] A recent hypothesis identifies the decrease of reelin signaling as the primary change in Alzheimer's disease that leads to the hyperphosphorylation of tau via a decrease in GSK3β inhibition. [65]

A68 is a name sometimes given (mostly in older publications) to the hyperphosphorylated form of tau protein found in the brains of individuals with Alzheimer's disease. [66]

In 2020, researchers from two groups published studies indicating that an immunoassay blood test for the p-tau-217 form of the protein could diagnose Alzheimer's up to decades before dementia symptoms were evident. [67] [68] [69]

Traumatic brain injury

Repetitive mild traumatic brain injury (TBI) is a central component of contact sports, especially American football, [70] [71] and the concussive force of military blasts. [72] It can lead to chronic traumatic encephalopathy (CTE), a condition characterized by fibrillar tangles of hyperphosphorylated tau. [73] After severe traumatic brain injury, high levels of tau protein in extracellular fluid in the brain are linked to poor outcomes. [74]

Prion-like propagation hypothesis

The term "prion-like" is often used to describe several aspects of tau pathology in various tauopathies, like Alzheimer's disease and frontotemporal dementia. [75] True prions are defined by their ability to induce misfolding of native proteins to perpetuate the pathology. True prions, like PRNP, are also infectious with the capability to cross species. Since tau has yet to be proven to be infectious it is not considered to be a true prion but instead a "prion-like" protein. Much like true prions, pathological tau aggregates have been shown to have the capacity to induce misfolding of native tau protein. [76] Both misfolding competent and non-misfolding competent species of tau aggregates have been reported, indicating a highly specific mechanism. [77]

Interactions

Tau protein has been shown to interact with:

See also

Related Research Articles

<span class="mw-page-title-main">Frontotemporal dementia</span> Types of dementia involving the frontal or temporal lobes

Frontotemporal dementia (FTD), frontotemporal degeneration disease, or frontotemporal neurocognitive disorder encompasses several types of dementia involving the progressive degeneration of the brain's frontal and temporal lobes. FTDs broadly present as behavioral or language disorders with gradual onsets.

In cell biology, microtubule-associated proteins (MAPs) are proteins that interact with the microtubules of the cellular cytoskeleton. MAPs are integral to the stability of the cell and its internal structures and the transport of components within the cell.

<span class="mw-page-title-main">Amyloid plaques</span> Extracellular deposits of the amyloid beta protein

Amyloid plaques are extracellular deposits of the amyloid beta (Aβ) protein mainly in the grey matter of the brain. Degenerative neuronal elements and an abundance of microglia and astrocytes can be associated with amyloid plaques. Some plaques occur in the brain as a result of aging, but large numbers of plaques and neurofibrillary tangles are characteristic features of Alzheimer's disease. The plaques are highly variable in shape and size; in tissue sections immunostained for Aβ, they comprise a log-normal size distribution curve, with an average plaque area of 400-450 square micrometers (µm²). The smallest plaques, which often consist of diffuse deposits of Aβ, are particularly numerous. Plaques form when Aβ misfolds and aggregates into oligomers and longer polymers, the latter of which are characteristic of amyloid.

<span class="mw-page-title-main">Frontotemporal lobar degeneration</span> Medical condition

Frontotemporal lobar degeneration (FTLD) is a pathological process that occurs in frontotemporal dementia. It is characterized by atrophy in the frontal lobe and temporal lobe of the brain, with sparing of the parietal and occipital lobes.

<span class="mw-page-title-main">Neurofibrillary tangle</span> Aggregates of tau protein known as a biomarker of Alzheimers disease

Neurofibrillary tangles (NFTs) are intracellular aggregates of hyperphosphorylated tau protein that are most commonly known as a primary biomarker of Alzheimer's disease. Their presence is also found in numerous other diseases known as tauopathies. Little is known about their exact relationship to the different pathologies.

<span class="mw-page-title-main">Corticobasal degeneration</span> Rare neurodegenerative disease

Corticobasal degeneration (CBD) is a rare neurodegenerative disease involving the cerebral cortex and the basal ganglia. CBD symptoms typically begin in people from 50 to 70 years of age, and typical survival before death is eight years. It is characterized by marked disorders in movement and cognition, and is classified as one of the Parkinson plus syndromes. Diagnosis is difficult, as symptoms are often similar to those of other disorders, such as Parkinson's disease, progressive supranuclear palsy, and dementia with Lewy bodies, and a definitive diagnosis of CBD can only be made upon neuropathologic examination.

<span class="mw-page-title-main">Major prion protein</span> Protein involved in multiple prion diseases

Major prion protein (PrP) is encoded in the human body by the PRNP gene also known as CD230. Expression of the protein is most predominant in the nervous system but occurs in many other tissues throughout the body.

<span class="mw-page-title-main">Tauopathy</span> Medical condition

Tauopathies are neurodegenerative diseases involving the aggregation of abnormal tau protein. Tangles are formed by hyperphosphorylation of the microtubule protein known as tau, causing the protein to dissociate from microtubules and form insoluble aggregate. Various neuropathologic phenotypes are identified based on the specific engagement of anatomical regions, cell types, and the presence of unique isoforms of tau within pathological deposits. The designation 'primary tauopathy' is assigned to disorders where the predominant feature is the deposition of tau protein. Alternatively, diseases exhibiting tau pathologies attributed to different and varied underlying causes are termed 'secondary tauopathies. Some neuropathologic phenotypes involving tau protein is Alzheimer's disease, Pick disease, Progressive supranuclear palsy and corticobasal degeneration.

<span class="mw-page-title-main">Neurodegenerative disease</span> Central nervous system disease

A neurodegenerative disease is caused by the progressive loss of structure or function of neurons, in the process known as neurodegeneration. Such neuronal damage may ultimately involve cell death. Neurodegenerative diseases include amyotrophic lateral sclerosis, multiple sclerosis, Parkinson's disease, Alzheimer's disease, Huntington's disease, multiple system atrophy, tauopathies, and prion diseases. Neurodegeneration can be found in the brain at many different levels of neuronal circuitry, ranging from molecular to systemic. Because there is no known way to reverse the progressive degeneration of neurons, these diseases are considered to be incurable; however research has shown that the two major contributing factors to neurodegeneration are oxidative stress and inflammation. Biomedical research has revealed many similarities between these diseases at the subcellular level, including atypical protein assemblies and induced cell death. These similarities suggest that therapeutic advances against one neurodegenerative disease might ameliorate other diseases as well.

<span class="mw-page-title-main">Frontotemporal dementia and parkinsonism linked to chromosome 17</span> Medical condition

Frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) is an autosomal dominant neurodegenerative tauopathy and Parkinson plus syndrome. FTDP-17 is caused by mutations in the MAPT gene located on the q arm of chromosome 17, and has three cardinal features: behavioral and personality changes, cognitive impairment, and motor symptoms. FTDP-17 was defined during the International Consensus Conference in Ann Arbor, Michigan, in 1996.

The biochemistry of Alzheimer's disease, the most common cause of dementia, is not yet very well understood. Alzheimer's disease (AD) has been identified as a proteopathy: a protein misfolding disease due to the accumulation of abnormally folded amyloid beta (Aβ) protein in the brain. Amyloid beta is a short peptide that is an abnormal proteolytic byproduct of the transmembrane protein amyloid-beta precursor protein (APP), whose function is unclear but thought to be involved in neuronal development. The presenilins are components of proteolytic complex involved in APP processing and degradation.

<span class="mw-page-title-main">Proteinopathy</span> Medical condition

In medicine, proteinopathy, or proteopathy, protein conformational disorder, or protein misfolding disease, is a class of diseases in which certain proteins become structurally abnormal, and thereby disrupt the function of cells, tissues and organs of the body. Often the proteins fail to fold into their normal configuration; in this misfolded state, the proteins can become toxic in some way or they can lose their normal function. The proteinopathies include such diseases as Creutzfeldt–Jakob disease and other prion diseases, Alzheimer's disease, Parkinson's disease, amyloidosis, multiple system atrophy, and a wide range of other disorders. The term proteopathy was first proposed in 2000 by Lary Walker and Harry LeVine.

<span class="mw-page-title-main">MAP4</span> Protein-coding gene in the species Homo sapiens

Microtubule-associated protein 4 is a protein that in humans is encoded by the MAP4 gene.

<span class="mw-page-title-main">MARK1</span> Protein-coding gene in the species Homo sapiens

Serine/threonine-protein kinase MARK1 is an enzyme that in humans is encoded by the MARK1 gene.

<span class="mw-page-title-main">PHKG1</span> Protein-coding gene in the species Homo sapiens

Phosphorylase b kinase gamma catalytic chain, skeletal muscle isoform is an enzyme that in humans is encoded by the PHKG1 gene.

<span class="mw-page-title-main">TMEM106B</span> Protein-coding gene in the species Homo sapiens

Transmembrane protein 106B is a protein that is encoded by the TMEM106B gene. It is found primarily within neurons and oligodendrocytes in the central nervous system with its subcellular location being in lysosomal membranes. TMEM106B helps facilitate important functions for maintaining a healthy lysosome, and therefore certain mutations and polymorphisms can lead to issues with proper lysosomal function. Lysosomes are in charge of clearing out mis-folded proteins and other debris, and thus, play an important role in neurodegenerative diseases that are driven by the accumulation of various mis-folded proteins and aggregates. Due to its impact on lysosomal function, TMEM106B has been investigated and found to be associated to multiple neurodegenerative diseases.

<span class="mw-page-title-main">Alzheimer's disease</span> Progressive neurodegenerative disease

Alzheimer's disease (AD) is a neurodegenerative disease that usually starts slowly and progressively worsens, and is the cause of 60–70% of cases of dementia. The most common early symptom is difficulty in remembering recent events. As the disease advances, symptoms can include problems with language, disorientation, mood swings, loss of motivation, self-neglect, and behavioral issues. As a person's condition declines, they often withdraw from family and society. Gradually, bodily functions are lost, ultimately leading to death. Although the speed of progression can vary, the average life expectancy following diagnosis is three to twelve years.

Maria Grazia Spillantini, is Professor of Molecular Neurology in the Department of Clinical Neurosciences at the University of Cambridge. She is most noted for identifying the protein alpha-synuclein as the major component of Lewy bodies, the characteristic protein deposit found in the brain in Parkinson's disease and dementia with Lewy bodies. She has also identified mutations in the MAPT gene as a heritable cause for frontotemporal dementia.

Primary age-related tauopathy (PART) is a neuropathological designation introduced in 2014 to describe the neurofibrillary tangles (NFT) that are commonly observed in the brains of normally aged and cognitively impaired individuals that can occur independently of the amyloid plaques of Alzheimer's disease (AD). The term and diagnostic criteria for PART were developed by a large group of neuropathologists, spearheaded by Drs. John F. Crary and Peter T. Nelson. Despite some controversy, the term PART has been widely adopted, with the consensus criteria cited over 1130 times as of April 2023 according to Google Scholar.

<span class="mw-page-title-main">Neurotubule</span>

Neurotubules are microtubules found in neurons in nervous tissues. Along with neurofilaments and microfilaments, they form the cytoskeleton of neurons. Neurotubules are undivided hollow cylinders that are made up of tubulin protein polymers and arrays parallel to the plasma membrane in neurons. Neurotubules have an outer diameter of about 23 nm and an inner diameter, also known as the central core, of about 12 nm. The wall of the neurotubules is about 5 nm in width. There is a non-opaque clear zone surrounding the neurotubule and it is about 40 nm in diameter. Like microtubules, neurotubules are greatly dynamic and the length of them can be adjusted by polymerization and depolymerization of tubulin.

References

  1. 1 2 3 ENSG00000276155, ENSG00000277956 GRCh38: Ensembl release 89: ENSG00000186868, ENSG00000276155, ENSG00000277956 - Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000018411 - Ensembl, May 2017
  3. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. 1 2 Weingarten MD, Lockwood AH, Hwo SY, Kirschner MW (May 1975). "A protein factor essential for microtubule assembly". Proceedings of the National Academy of Sciences of the United States of America. 72 (5): 1858–62. Bibcode:1975PNAS...72.1858W. doi: 10.1073/pnas.72.5.1858 . PMC   432646 . PMID   1057175.
  6. Goedert M, Wischik CM, Crowther RA, Walker JE, Klug A (June 1988). "Cloning and sequencing of the cDNA encoding a core protein of the paired helical filament of Alzheimer disease: identification as the microtubule-associated protein tau". Proceedings of the National Academy of Sciences of the United States of America. 85 (11): 4051–5. Bibcode:1988PNAS...85.4051G. doi: 10.1073/pnas.85.11.4051 . PMC   280359 . PMID   3131773.
  7. Goedert M, Spillantini MG, Jakes R, Rutherford D, Crowther RA (October 1989). "Multiple isoforms of human microtubule-associated protein tau: sequences and localization in neurofibrillary tangles of Alzheimer's disease". Neuron. 3 (4): 519–26. doi:10.1016/0896-6273(89)90210-9. PMID   2484340. S2CID   19627629.
  8. Sjölin, Karl; Kultima, Kim; Larsson, Anders; Freyhult, Eva; Zjukovskaja, Christina; Alkass, Kanar; Burman, Joachim (December 2022). "Distribution of five clinically important neuroglial proteins in the human brain". Molecular Brain. 15 (1): 52. doi: 10.1186/s13041-022-00935-6 . ISSN   1756-6606. PMC   9241296 . PMID   35765081.
  9. Shin RW, Iwaki T, Kitamoto T, Tateishi J (May 1991). "Hydrated autoclave pretreatment enhances tau immunoreactivity in formalin-fixed normal and Alzheimer's disease brain tissues". Laboratory Investigation; A Journal of Technical Methods and Pathology. 64 (5): 693–702. PMID   1903170.
  10. Lei P, Ayton S, Finkelstein DI, Adlard PA, Masters CL, Bush AI (November 2010). "Tau protein: relevance to Parkinson's disease". The International Journal of Biochemistry & Cell Biology. 42 (11): 1775–8. doi:10.1016/j.biocel.2010.07.016. PMID   20678581.
  11. 1 2 3 Cleveland DW, Hwo SY, Kirschner MW (October 1977). "Purification of tau, a microtubule-associated protein that induces assembly of microtubules from purified tubulin". Journal of Molecular Biology. 116 (2): 207–25. doi:10.1016/0022-2836(77)90213-3. PMID   599557.
  12. Cleveland DW, Hwo SY, Kirschner MW (October 1977). "Physical and chemical properties of purified tau factor and the role of tau in microtubule assembly". Journal of Molecular Biology. 116 (2): 227–47. doi:10.1016/0022-2836(77)90214-5. PMID   146092.
  13. 1 2 3 4 5 6 Papanikolopoulou K, Roussou IG, Gouzi JY, Samiotaki M, Panayotou G, Turin L, Skoulakis EM (October 2019). "Drosophila Tau Negatively Regulates Translation and Olfactory Long-Term Memory, But Facilitates Footshock Habituation and Cytoskeletal Homeostasis". The Journal of Neuroscience. 39 (42): 8315–8329. doi:10.1523/JNEUROSCI.0391-19.2019. PMC   6794924 . PMID   31488613.
  14. 1 2 Harada A, Oguchi K, Okabe S, Kuno J, Terada S, Ohshima T, et al. (June 1994). "Altered microtubule organization in small-calibre axons of mice lacking tau protein". Nature. 369 (6480): 488–91. Bibcode:1994Natur.369..488H. doi:10.1038/369488a0. PMID   8202139. S2CID   4322543.
  15. 1 2 Wang JZ, Liu F (June 2008). "Microtubule-associated protein tau in development, degeneration and protection of neurons". Progress in Neurobiology. 85 (2): 148–75. doi:10.1016/j.pneurobio.2008.03.002. PMID   18448228. S2CID   32708424.
  16. Ke YD, Suchowerska AK, van der Hoven J, De Silva DM, Wu CW, van Eersel J, et al. (June 2012). "Lessons from tau-deficient mice". International Journal of Alzheimer's Disease. 2012 (873270): 873270. doi: 10.1155/2012/873270 . PMC   3375147 . PMID   22720190.
  17. Ittner LM, Ke YD, Delerue F, Bi M, Gladbach A, van Eersel J, et al. (August 2010). "Dendritic function of tau mediates amyloid-beta toxicity in Alzheimer's disease mouse models". Cell. 142 (3): 387–97. doi: 10.1016/j.cell.2010.06.036 . PMID   20655099. S2CID   18776289.
  18. Dehmelt L, Halpain S (2004). "The MAP2/Tau family of microtubule-associated proteins". Genome Biology. 6 (1): 204. doi: 10.1186/gb-2004-6-1-204 . PMC   549057 . PMID   15642108.
  19. Evans HT, Benetatos J, van Roigen M, Bodea LG, Götz J (2019-07-01). "Decreased synthesis of ribosomal proteins in tauopathy revealed by non-canonical amino acid labelling". The EMBO Journal. 38 (13): e101174. doi:10.15252/embj.2018101174. ISSN   0261-4189. PMC   6600635 . PMID   31268600.
  20. 1 2 Meier S, Bell M, Lyons DN, Rodriguez-Rivera J, Ingram A, Fontaine SN, et al. (January 2016). "Pathological Tau Promotes Neuronal Damage by Impairing Ribosomal Function and Decreasing Protein Synthesis". The Journal of Neuroscience. 36 (3): 1001–7. doi:10.1523/JNEUROSCI.3029-15.2016. PMC   4719006 . PMID   26791227.
  21. Evans HT, Taylor D, Kneynsberg A, Bodea LG, Götz J (2021-06-19). "Altered ribosomal function and protein synthesis caused by tau". Acta Neuropathologica Communications. 9 (1): 110. doi: 10.1186/s40478-021-01208-4 . ISSN   2051-5960. PMC   8214309 . PMID   34147135.
  22. Koren SA, Hamm MJ, Meier SE, Weiss BE, Nation GK, Chishti EA, et al. (April 2019). "Tau drives translational selectivity by interacting with ribosomal proteins". Acta Neuropathologica. 137 (4): 571–583. doi:10.1007/s00401-019-01970-9. PMC   6426815 . PMID   30759285.
  23. Dawson HN, Ferreira A, Eyster MV, Ghoshal N, Binder LI, Vitek MP (March 2001). "Inhibition of neuronal maturation in primary hippocampal neurons from tau deficient mice". Journal of Cell Science. 114 (Pt 6): 1179–87. doi:10.1242/jcs.114.6.1179. PMID   11228161.
  24. Fujio K, Sato M, Uemura T, Sato T, Sato-Harada R, Harada A (July 2007). "14-3-3 proteins and protein phosphatases are not reduced in tau-deficient mice". NeuroReport. 18 (10): 1049–52. doi:10.1097/WNR.0b013e32818b2a0b. PMID   17558294. S2CID   25235996.
  25. 1 2 Ikegami S, Harada A, Hirokawa N (February 2000). "Muscle weakness, hyperactivity, and impairment in fear conditioning in tau-deficient mice". Neuroscience Letters. 279 (3): 129–32. doi:10.1016/s0304-3940(99)00964-7. PMID   10688046. S2CID   31204860.
  26. Roberson ED, Scearce-Levie K, Palop JJ, Yan F, Cheng IH, Wu T, et al. (May 2007). "Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer's disease mouse model". Science. 316 (5825): 750–4. Bibcode:2007Sci...316..750R. doi:10.1126/science.1141736. PMID   17478722. S2CID   32771613.
  27. Dawson HN, Cantillana V, Jansen M, Wang H, Vitek MP, Wilcock DM, et al. (August 2010). "Loss of tau elicits axonal degeneration in a mouse model of Alzheimer's disease". Neuroscience. 169 (1): 516–31. doi:10.1016/j.neuroscience.2010.04.037. PMC   2900546 . PMID   20434528.
  28. Cantero JL, Hita-Yañez E, Moreno-Lopez B, Portillo F, Rubio A, Avila J (August 2010). "Tau protein role in sleep-wake cycle". Journal of Alzheimer's Disease. 21 (2): 411–21. doi:10.3233/JAD-2010-100285. PMID   20555133.
  29. Sotiropoulos I, Galas MC, Silva JM, Skoulakis E, Wegmann S, Maina MB, et al. (November 2017). "Atypical, non-standard functions of the microtubule associated Tau protein". Acta Neuropathologica Communications. 5 (1): 91. doi: 10.1186/s40478-017-0489-6 . PMC   5707803 . PMID   29187252.
  30. Neve RL, Harris P, Kosik KS, Kurnit DM, Donlon TA (December 1986). "Identification of cDNA clones for the human microtubule-associated protein tau and chromosomal localization of the genes for tau and microtubule-associated protein 2". Brain Research. 387 (3): 271–80. doi:10.1016/0169-328x(86)90033-1. PMID   3103857.
  31. Sergeant N, Delacourte A, Buée L (January 2005). "Tau protein as a differential biomarker of tauopathies". Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 1739 (2–3): 179–97. doi: 10.1016/j.bbadis.2004.06.020 . PMID   15615637.
  32. Shaw-Smith C, Pittman AM, Willatt L, Martin H, Rickman L, Gribble S, et al. (September 2006). "Microdeletion encompassing MAPT at chromosome 17q21.3 is associated with developmental delay and learning disability". Nature Genetics. 38 (9): 1032–7. doi:10.1038/ng1858. PMID   16906163. S2CID   38047848.
  33. Zody MC, Jiang Z, Fung HC, Antonacci F, Hillier LW, Cardone MF, et al. (September 2008). "Evolutionary toggling of the MAPT 17q21.31 inversion region". Nature Genetics. 40 (9): 1076–83. doi:10.1038/ng.193. PMC   2684794 . PMID   19165922.
  34. Almos PZ, Horváth S, Czibula A, Raskó I, Sipos B, Bihari P, et al. (November 2008). "H1 tau haplotype-related genomic variation at 17q21.3 as an Asian heritage of the European Gypsy population". Heredity. 101 (5): 416–9. doi: 10.1038/hdy.2008.70 . PMID   18648385.
  35. Hardy J, Pittman A, Myers A, Gwinn-Hardy K, Fung HC, de Silva R, et al. (August 2005). "Evidence suggesting that Homo neanderthalensis contributed the H2 MAPT haplotype to Homo sapiens". Biochemical Society Transactions. 33 (Pt 4): 582–5. doi:10.1042/BST0330582. PMID   16042549.
  36. Billingsley ML, Kincaid RL (May 1997). "Regulated phosphorylation and dephosphorylation of tau protein: effects on microtubule interaction, intracellular trafficking and neurodegeneration". The Biochemical Journal. 323 ( Pt 3) (3): 577–91. doi:10.1042/bj3230577. PMC   1218358 . PMID   9169588.
  37. Taniguchi T, Kawamata T, Mukai H, Hasegawa H, Isagawa T, Yasuda M, et al. (March 2001). "Phosphorylation of tau is regulated by PKN". The Journal of Biological Chemistry. 276 (13): 10025–31. doi: 10.1074/jbc.M007427200 . PMID   11104762.
  38. Kanemaru K, Takio K, Miura R, Titani K, Ihara Y (May 1992). "Fetal-type phosphorylation of the tau in paired helical filaments". Journal of Neurochemistry. 58 (5): 1667–75. doi:10.1111/j.1471-4159.1992.tb10039.x. PMID   1560225. S2CID   94265621.
  39. Mawal-Dewan M, Henley J, Van de Voorde A, Trojanowski JQ, Lee VM (December 1994). "The phosphorylation state of tau in the developing rat brain is regulated by phosphoprotein phosphatases". The Journal of Biological Chemistry. 269 (49): 30981–7. doi: 10.1016/S0021-9258(18)47378-4 . PMID   7983034.
  40. Matsuo ES, Shin RW, Billingsley ML, Van deVoorde A, O'Connor M, Trojanowski JQ, Lee VM (October 1994). "Biopsy-derived adult human brain tau is phosphorylated at many of the same sites as Alzheimer's disease paired helical filament tau". Neuron. 13 (4): 989–1002. doi:10.1016/0896-6273(94)90264-X. PMID   7946342. S2CID   40592137.
  41. Liu, Fei; Iqbal, Khalid; Grundke-Iqbal, Inge; Hart, Gerald W.; Gong, Cheng-Xin (2004-07-20). "O-GlcNAcylation regulates phosphorylation of tau: A mechanism involved in Alzheimer's disease". Proceedings of the National Academy of Sciences of the United States of America. 101 (29): 10804–10809. Bibcode:2004PNAS..10110804L. doi: 10.1073/pnas.0400348101 . ISSN   0027-8424. PMC   490015 . PMID   15249677.
  42. Alonso AD, Grundke-Iqbal I, Barra HS, Iqbal K (January 1997). "Abnormal phosphorylation of tau and the mechanism of Alzheimer neurofibrillary degeneration: sequestration of microtubule-associated proteins 1 and 2 and the disassembly of microtubules by the abnormal tau". Proceedings of the National Academy of Sciences of the United States of America. 94 (1): 298–303. Bibcode:1997PNAS...94..298A. doi: 10.1073/pnas.94.1.298 . PMC   19321 . PMID   8990203.
  43. Frost B, Jacks RL, Diamond MI (May 2009). "Propagation of tau misfolding from the outside to the inside of a cell". The Journal of Biological Chemistry. 284 (19): 12845–52. doi: 10.1074/jbc.M808759200 . PMC   2676015 . PMID   19282288.
  44. Calafate S, Buist A, Miskiewicz K, Vijayan V, Daneels G, de Strooper B, et al. (May 2015). "Synaptic Contacts Enhance Cell-to-Cell Tau Pathology Propagation" (PDF). Cell Reports. 11 (8): 1176–83. doi: 10.1016/j.celrep.2015.04.043 . PMID   25981034.
  45. Roman AY, Devred F, Byrne D, La Rocca R, Ninkina NN, Peyrot V, Tsvetkov PO (February 2019). "Zinc Induces Temperature-Dependent Reversible Self-Assembly of Tau". Journal of Molecular Biology. 431 (4): 687–695. doi: 10.1016/j.jmb.2018.12.008 . PMID   30580037.
  46. Fichou Y, Al-Hilaly YK, Devred F, Smet-Nocca C, Tsvetkov PO, Verelst J, et al. (March 2019). "The elusive tau molecular structures: can we translate the recent breakthroughs into new targets for intervention?". Acta Neuropathologica Communications. 7 (1): 31. doi: 10.1186/s40478-019-0682-x . PMC   6397507 . PMID   30823892.
  47. Goedert M, Eisenberg DS, Crowther RA (July 2017). "Propagation of Tau Aggregates and Neurodegeneration". Annual Review of Neuroscience. 40 (1): 189–210. doi: 10.1146/annurev-neuro-072116-031153 . PMID   28772101.
  48. Yamada K (2017). "Extracellular Tau and Its Potential Role in the Propagation of Tau Pathology". Frontiers in Neuroscience. 11: 667. doi: 10.3389/fnins.2017.00667 . PMC   5712583 . PMID   29238289.
  49. Asai H, Ikezu S, Tsunoda S, Medalla M, Luebke J, Haydar T, et al. (November 2015). "Depletion of microglia and inhibition of exosome synthesis halt tau propagation". Nature Neuroscience. 18 (11): 1584–93. doi:10.1038/nn.4132. PMC   4694577 . PMID   26436904.
  50. Nishimura I, Yang Y, Lu B (March 2004). "PAR-1 kinase plays an initiator role in a temporally ordered phosphorylation process that confers tau toxicity in Drosophila". Cell. 116 (5): 671–82. doi: 10.1016/S0092-8674(04)00170-9 . PMID   15006350. S2CID   18896805.
  51. Chatterjee S, Sang TK, Lawless GM, Jackson GR (January 2009). "Dissociation of tau toxicity and phosphorylation: role of GSK-3beta, MARK and Cdk5 in a Drosophila model". Human Molecular Genetics. 18 (1): 164–77. doi:10.1093/hmg/ddn326. PMC   2644648 . PMID   18930955.
  52. Lee HG, Perry G, Moreira PI, Garrett MR, Liu Q, Zhu X, et al. (April 2005). "Tau phosphorylation in Alzheimer's disease: pathogen or protector?". Trends in Molecular Medicine. 11 (4): 164–9. doi:10.1016/j.molmed.2005.02.008. hdl: 10316/4769 . PMID   15823754.
  53. Alonso A, Zaidi T, Novak M, Grundke-Iqbal I, Iqbal K (June 2001). "Hyperphosphorylation induces self-assembly of tau into tangles of paired helical filaments/straight filaments". Proceedings of the National Academy of Sciences of the United States of America. 98 (12): 6923–8. Bibcode:2001PNAS...98.6923A. doi: 10.1073/pnas.121119298 . PMC   34454 . PMID   11381127.
  54. 1 2 "Alzheimer's Brain Tangles." Alzheimer's Association, www.alz.org/braintour/tangles.asp.
  55. Köglsberger S, Cordero-Maldonado ML, Antony P, Forster JI, Garcia P, Buttini M, et al. (December 2017). "Gender-Specific Expression of Ubiquitin-Specific Peptidase 9 Modulates Tau Expression and Phosphorylation: Possible Implications for Tauopathies". Molecular Neurobiology. 54 (10): 7979–7993. doi:10.1007/s12035-016-0299-z. PMC   5684262 . PMID   27878758.
  56. Hall GF, Patuto BA (July 2012). "Is tau ready for admission to the prion club?". Prion. 6 (3): 223–33. doi:10.4161/pri.19912. PMC   3399531 . PMID   22561167.
  57. Mohandas E, Rajmohan V, Raghunath B (January 2009). "Neurobiology of Alzheimer's disease". Indian Journal of Psychiatry. 51 (1): 55–61. doi: 10.4103/0019-5545.44908 . PMC   2738403 . PMID   19742193.
  58. Augustinack JC, Schneider A, Mandelkow EM, Hyman BT (January 2002). "Specific tau phosphorylation sites correlate with severity of neuronal cytopathology in Alzheimer's disease". Acta Neuropathologica. 103 (1): 26–35. doi:10.1007/s004010100423. PMID   11837744. S2CID   6799990.
  59. van Slegtenhorst M, Lewis J, Hutton M (July 2000). "The molecular genetics of the tauopathies". Experimental Gerontology. 35 (4): 461–71. doi:10.1016/S0531-5565(00)00114-5. PMID   10959034. S2CID   38730940.
  60. Mudher A, Lovestone S (January 2002). "Alzheimer's disease-do tauists and baptists finally shake hands?". Trends in Neurosciences. 25 (1): 22–6. doi:10.1016/s0166-2236(00)02031-2. PMID   11801334. S2CID   37380445.
  61. Pradeepkiran JA, Reddy PH (March 2019). "Structure Based Design and Molecular Docking Studies for Phosphorylated Tau Inhibitors in Alzheimer's Disease". Cells. 8 (3): 260. doi: 10.3390/cells8030260 . PMC   6468864 . PMID   30893872.
  62. Tepper K, Biernat J, Kumar S, Wegmann S, Timm T, Hübschmann S, et al. (December 2014). "Oligomer formation of tau protein hyperphosphorylated in cells". The Journal of Biological Chemistry. 289 (49): 34389–407. doi: 10.1074/jbc.M114.611368 . PMC   4256367 . PMID   25339173.
  63. Shin RW, Bramblett GT, Lee VM, Trojanowski JQ (July 1993). "Alzheimer disease A68 proteins injected into rat brain induce codeposits of beta-amyloid, ubiquitin, and alpha 1-antichymotrypsin". Proceedings of the National Academy of Sciences of the United States of America. 90 (14): 6825–8. Bibcode:1993PNAS...90.6825S. doi: 10.1073/pnas.90.14.6825 . PMC   47025 . PMID   8393578.
  64. Vincent IJ, Davies P (October 1990). "Phosphorylation characteristics of the A68 protein in Alzheimer's disease". Brain Research. 531 (1–2): 127–35. doi:10.1016/0006-8993(90)90765-4. PMID   2126970. S2CID   23900723.
  65. Kovács KA (December 2021). "Relevance of a Novel Circuit-Level Model of Episodic Memories to Alzheimer's Disease". International Journal of Molecular Sciences. 23 (1): 462. doi: 10.3390/ijms23010462 . PMC   8745479 . PMID   35008886.
  66. "A68", The Free Dictionary, retrieved 2020-01-27
  67. Belluck, Pam (2020-07-28). "'Amazing, Isn't It?' Long-Sought Blood Test for Alzheimer's in Reach". The New York Times. ISSN   0362-4331 . Retrieved 2020-07-29.
  68. Barthélemy, Nicolas R.; Horie, Kanta; Sato, Chihiro; Bateman, Randall J. (2020-11-02). "Blood plasma phosphorylated-tau isoforms track CNS change in Alzheimer's disease". Journal of Experimental Medicine. 217 (11). doi: 10.1084/jem.20200861 . ISSN   0022-1007. PMC   7596823 . PMID   32725127.
  69. Palmqvist, Sebastian; Janelidze, Shorena; Quiroz, Yakeel T.; Zetterberg, Henrik; Lopera, Francisco; Stomrud, Erik; Su, Yi; Chen, Yinghua; Serrano, Geidy E.; Leuzy, Antoine; Mattsson-Carlgren, Niklas (2020-07-28). "Discriminative Accuracy of Plasma Phospho-tau217 for Alzheimer Disease vs Other Neurodegenerative Disorders". JAMA. 324 (8): 772–781. doi:10.1001/jama.2020.12134. PMC   7388060 . PMID   32722745.
  70. "Brain Trauma". NOVA. PBS Online by WGBH.
  71. Omalu BI, DeKosky ST, Minster RL, Kamboh MI, Hamilton RL, Wecht CH (July 2005). "Chronic traumatic encephalopathy in a National Football League player". Neurosurgery. 57 (1): 128–34, discussion 128–34. doi:10.1227/01.NEU.0000163407.92769.ED. PMID   15987548. S2CID   196391183.
  72. Goldstein LE, Fisher AM, Tagge CA, Zhang XL, Velisek L, Sullivan JA, et al. (May 2012). "Chronic traumatic encephalopathy in blast-exposed military veterans and a blast neurotrauma mouse model". Science Translational Medicine. 4 (134): 134ra60. doi:10.1126/scitranslmed.3003716. PMC   3739428 . PMID   22593173.
  73. McKee AC, Stern RA, Nowinski CJ, Stein TD, Alvarez VE, Daneshvar DH, et al. (January 2013). "The spectrum of disease in chronic traumatic encephalopathy". Brain. 136 (Pt 1): 43–64. doi:10.1093/brain/aws307. PMC   3624697 . PMID   23208308.
  74. Magnoni S, Esparza TJ, Conte V, Carbonara M, Carrabba G, Holtzman DM, et al. (April 2012). "Tau elevations in the brain extracellular space correlate with reduced amyloid-β levels and predict adverse clinical outcomes after severe traumatic brain injury". Brain. 135 (Pt 4): 1268–80. doi:10.1093/brain/awr286. PMC   3326246 . PMID   22116192.
  75. Dujardin, Simon; Hyman, Bradley T. (2019), Takashima, Akihiko; Wolozin, Benjamin; Buee, Luc (eds.), "Tau Prion-Like Propagation: State of the Art and Current Challenges", Tau Biology, vol. 1184, Singapore: Springer Singapore, pp. 305–325, doi:10.1007/978-981-32-9358-8_23, ISBN   978-981-329-357-1, PMID   32096046, S2CID   211475911 , retrieved 2021-03-18
  76. Bloom, George S. (2014-04-01). "Amyloid-β and Tau: The Trigger and Bullet in Alzheimer Disease Pathogenesis". JAMA Neurology. 71 (4): 505–508. doi: 10.1001/jamaneurol.2013.5847 . ISSN   2168-6149. PMID   24493463.
  77. Hosokawa, Masato; Hasegawa, Masato (2019-07-01). "Prion-like Propagation Model of Tau". Yakugaku Zasshi. 139 (7): 1021–1025. doi: 10.1248/yakushi.18-00165-6 . ISSN   0031-6903. PMID   31257249.
  78. Jensen PH, Hager H, Nielsen MS, Hojrup P, Gliemann J, Jakes R (September 1999). "alpha-synuclein binds to Tau and stimulates the protein kinase A-catalyzed tau phosphorylation of serine residues 262 and 356". The Journal of Biological Chemistry. 274 (36): 25481–9. doi: 10.1074/jbc.274.36.25481 . PMID   10464279.
  79. Giasson BI, Lee VM, Trojanowski JQ (2003). "Interactions of amyloidogenic proteins". Neuromolecular Medicine. 4 (1–2): 49–58. doi:10.1385/NMM:4:1-2:49. PMID   14528052. S2CID   9086733.
  80. Klein C, Kramer EM, Cardine AM, Schraven B, Brandt R, Trotter J (February 2002). "Process outgrowth of oligodendrocytes is promoted by interaction of fyn kinase with the cytoskeletal protein tau". The Journal of Neuroscience. 22 (3): 698–707. doi:10.1523/JNEUROSCI.22-03-00698.2002. PMC   6758498 . PMID   11826099.
  81. Yu WH, Fraser PE (April 2001). "S100beta interaction with tau is promoted by zinc and inhibited by hyperphosphorylation in Alzheimer's disease". The Journal of Neuroscience. 21 (7): 2240–6. doi:10.1523/JNEUROSCI.21-07-02240.2001. PMC   6762409 . PMID   11264299.
  82. Baudier J, Cole RD (April 1988). "Interactions between the microtubule-associated tau proteins and S100b regulate tau phosphorylation by the Ca2+/calmodulin-dependent protein kinase II". The Journal of Biological Chemistry. 263 (12): 5876–83. doi: 10.1016/S0021-9258(18)60647-7 . PMID   2833519.
  83. Hashiguchi M, Sobue K, Paudel HK (August 2000). "14-3-3zeta is an effector of tau protein phosphorylation". The Journal of Biological Chemistry. 275 (33): 25247–54. doi: 10.1074/jbc.M003738200 . PMID   10840038.

Further reading