Elsevier

Biochemical Pharmacology

Volume 88, Issue 4, 15 April 2014, Pages 540-547
Biochemical Pharmacology

Review - Part of the Special Issue: Alzheimer's Disease - Amyloid, Tau and Beyond
New perspectives on the role of tau in Alzheimer's disease. Implications for therapy

https://doi.org/10.1016/j.bcp.2014.01.013Get rights and content

Abstract

Alzheimer's disease (AD) and related dementias constitute a major public health issue due to an increasingly aged population as a consequence of generally improved medical care and demographic changes. Current drug treatment of AD, the most prevalent dementia, with cholinesterase inhibitors or NMDA antagonists have demonstrated very modest, symptomatic efficacy, leaving an unmet medical need for new, more effective therapies. While drug development efforts in the last two decades have primarily focused on the amyloid cascade hypothesis, so far with disappointing results, tau-based strategies have received little attention until recently despite that the presence of extensive tau pathology is central to the disease. The discovery of mutations within the tau gene that cause fronto-temporal dementia demonstrated that tau dysfunction, in the absence of amyloid pathology, was sufficient to cause neuronal loss and clinical dementia. Abnormal levels and hyperphosphorylation of tau protein have been reported to be the underlying cause of a group of neurodegenerative disorders collectively known as ‘tauopathies’. The detrimental consequence is the loss of affinity between this protein and the microtubules, increased production of fibrillary aggregates and the accumulation of insoluble intracellular neurofibrillary tangles. However, it has become clear in recent years that tau is not only a microtubule interacting protein, but rather has additional roles in cellular processes. This review focuses on emerging therapeutic strategies aimed at treating the underlying causes of the tau pathology in tauopathies and AD, including some novel approaches on the verge of providing new treatment paradigms within the coming years.

Introduction

Tau protein is a microtubule-associated protein (MAP) that under physiological conditions regulates microtubules (MT) assembly, dynamic behavior, and spatial organization [1], [2]. More recently, tau has been shown to regulate the axonal transport of organelles, including mitochondria [3]. Tau is encoded by a single gene from which six major isoforms are expressed in adult brain through alternative splicing [4], [5]. The interaction between tau and tubulin is mediated by four imperfect repeat domains (encompassing 31–32 residues) encoded by exons 9–12 [6]. Alternative splicing of exon 10 results in the production of isoforms containing 3 or 4 binding domains (3R and 4R tau) [5]. Adult human brain contains equal amounts of 3R and 4R isoforms whereas foetal brain, however, only expresses 3R tau, demonstrating developmental regulation of exon 10 splicing [7]. Different brain regions also differ in the relative levels of 3R and 4R isoforms with granule cells in the hippocampal formation reported to have only 3R tau [5].

Within neurons, tau is predominantly found in axons [8] as a highly soluble phosphoprotein [9]. Phosphorylation is also developmentally regulated, with a high tau phosphorylation level during embryogenesis and early development, when only the shortest of the isoforms is being expressed. By contrast, adult brain expresses all six isoforms with relatively reduced phosphorylation levels compared with the foetal one (see [10] for a review).

A number of neurodegenerative disorders, including Alzheimer's disease (AD), present prominent tau pathology in the CNS, collectively referred to as tauopathies [11], [12], [13], [14], [15]. In tauopathies, the natively unfolded tau protein forms intracellular fibrillar structures of aggregated, hyperphosphorylated, and ubiquinated tau, which are associated with synaptic and neuronal loss. Upon abnormal phosphorylation, the microtubule-associated protein tau reduces its affinity for and dissociates from microtubules[15], [16], [17] In Alzheimer's disease (AD) brains tau accumulates in the neuronal perikarya and processes as paired helical filaments (PHF) [18], [19]. It has been suggested that at the single-cell level the defects start with a modification of tau by phosphorylation, resulting in a destabilization of microtubules giving rise to a “pre-tangle” stage [20]. After this stage, the destabilization of microtubules leads to loss of dendritic microtubules and synapses, plasma membrane degeneration, and eventually cell death [21].

The occurrence of fibrillar tau inclusions in tauopathies strongly supports a key role in the observed clinical symptoms and pathology. As a matter of fact, tau pathology in AD correlates better than amyloid-β (Aβ) pathology with the cognitive impairment observed in patients [22]. Furthermore, the discovery of mutations within the tau gene that cause a disease known as fronto-temporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) [23], demonstrated that tau dysfunction, in the absence of amyloid pathology (as occurs in AD), was sufficient to cause neuronal loss and clinical dementia. Interestingly, tau is needed for Aβ to cause its toxicity both in vitro and in vivo, as suggested by the resistance shown by primary cultured neurons from tau knockout mice when exposed to Aβ [24] as well as by the rescuing premature mortality and preventing memory deficits observed in APP transgenic mice when crossed with tau knockout mice [25]. This supports a central role of tau in mediating Aβ toxicity in the early pathogenesis of AD.

Taken together, there is a large body of evidence clearly pointing out to a central role for tau in the pathogenesis of several neurodegenerative conditions including AD, laying the groundwork for its use as a target for the development of novel disease modifying therapeutics [26]. Drug discovery and development efforts for AD in the last two decades have primarily focused on targets defined by the amyloid cascade hypothesis, so far with disappointing results. In contrast, despite the fact that the presence of extensive tau pathology is central to the disease, tau-based strategies have received little attention until recently [27].

Section snippets

Tau-induced neurodegeneration

The knowledge accumulated in recent years strongly suggest that tau-induced neurodegeneration is most likely a consequence of a combination of loss of (tau) function as well as gain of (toxic) functions. On one hand, tau detachment from microtubules after hyperphosphorylation (or mutations) causes impaired microtubule function and axonal transport and eventually synaptic dysfunction and neuronal loss [28]. On the other hand, hyperphosphorylated tau molecules tend to self-assemble into filaments

Modulating tau phosphorylation

Although it is still debated whether phosphorylation of tau drives aggregation, or is a consequence of it, the strong correlation that exists between phosphorylation and tau pathology has laid the foundation to look for tau kinase inhibitors as potential therapeutics. However, there is not a single but multiple kinases involved in generating hyperphosphorylated tau in vivo [10], raising the issue of whether targeting specific kinases or distinct groups of kinases will be more effective in

Microtubule stabilization

Based on the notion that tau detachment from microtubules results in loss of its normal microtubule stabilizing function, most likely leading to axonal transport impairment and eventually to synaptic dysfunction, some microtubule-stabilizing agents have also been proposed as a therapeutic alternative. Among those, some anti-mitotic compounds such as paclitaxel or epothilone have been used in tau transgenic animals (reviewed in [49]). The most advanced of these agents is davunetide (NAP), an

Inhibiting tau aggregation

Another potential point of therapeutic intervention is the aggregation process itself in which a variety of different tau species are formed: monomers, oligomers, prefilaments, truncated, granules, fibrils, insoluble aggregates. As discussed above, some sort of hyperphosphorylated, misfolded soluble tau intermediates are the actual (or the most) toxic tau species, so that inhibiting early aggregation steps might be critical for a successful pharmacological intervention aimed at preventing

Reducing tau levels

It has been over twenty years that increased intracellular tau levels were described in brains from AD patients as compared to age-matched non-demented controls [58] and it has thus been suggested that increased tau levels might be toxic for neurons. On the other hand, several tau-deficient mouse strains have been generated [61], [62], [63], [64], [65], [66], [67] and while initially reported to lack an overt phenotype, behavioral changes and motor deficits have been reported recently in some

Decreasing extracellular tau

Development of tau pathology is associated with progressive neuronal loss and cognitive decline. In the brains of AD patients, tau pathology spreads following a predictable, anatomically defined progression pattern, which has suggested that the actual interneuronal transfer of a toxic factor must be involved in the progression of neurodegenerative tauopathies [84], [85]. Although traditionally thought of as an exclusively intracellular protein and its presence in the extracellular milieu (or in

Immunotherapy targeting tau

Immunotherapy for various neurodegenerative diseases have recently emerged as a promising approach for the clearance of pathological proteins in these disorders [100]. The disappointing outcome of several amyloid-based pharmacological approaches, including anti-amyloid immunotherapy, in late stage clinical trials during the last few years has spurred increased interest in novel tau-based therapies and various vaccination approaches have been recently assessed in preclinical models for

Future developments

Drug discovery and development efforts for AD in the last two decades have primarily focused on targets defined by the amyloid cascade hypothesis, so far with disappointing results. In contrast, tau-based strategies have received little attention until recently despite that the presence of extensive tau pathology is central to the disease. However, as discussed in this review, a number of different strategies targeting several aspects of the tau pathogenesis are being actively pursued,

References (122)

  • A. Schneider et al.

    Tau-based treatment strategies in neurodegenerative diseases

    Neurotherapeutics

    (2008)
  • J.M. Domínguez et al.

    Evidence for irreversible inhibition of glycogen synthase kinase-3β by tideglusib

    J Biol Chem

    (2012)
  • T.E. Golde et al.

    Abeta and tau in Alzheimer's disease: an early interim report

    Exp Neurol

    (2010)
  • N. Shiryaev et al.

    NAP protects memory: increases soluble tau and reduces tau hyperphosphorylation in a tauopathy model

    Neurobiol Dis

    (2009)
  • C.M. Wischik et al.

    Tau aggregation inhibitor (TAI) therapy with Rember™ arrests disease progression in mild and moderate Alzheimer's disease over 50 weeks

    Alz Dement

    (2008)
  • C. Wischik et al.

    Challenges in the conduct of disease-modifying trials in AD: practical experience from a phase 2 trial of Tau-aggregation inhibitor therapy

    J Nutr Health Aging

    (2009)
  • L.M. Ittner et al.

    Dendritic function of tau mediates amyloid-β toxicity in Alzheimer's disease mouse models

    Cell

    (2010)
  • H.N. Dawson et al.

    Loss of tau elicits axonal degeneration in a mouse model of Alzheimer's disease

    Neuroscience

    (2010)
  • A.J. Myers et al.

    The MAPT H1c risk haplotype is associated with increased expression of tau and especially of 4 repeat containing transcripts

    Neurobiol Dis

    (2007)
  • M.J. Lee et al.

    Tau degradation: the ubiquitin-proteasome system versus the autophagy-lysosome system

    Prog Neurobiol

    (2013)
  • C.B. Saper et al.

    Axonal and transneuronal transport in the transmission of neurological disease: potential role in system degenerations, including Alzheimer's disease

    Neuroscience

    (1987)
  • J.H. Su et al.

    Transneuronal degeneration in the spread of Alzheimer's disease pathology: immunohistochemical evidence for the transmission of tau hyperphosphorylation

    Neurobiol Dis

    (1997)
  • C.M. Karch et al.

    Extracellular Tau levels are influenced by variability in Tau that is associated with tauopathies

    J Biol Chem

    (2012)
  • X. Chai et al.

    Constitutive secretion of tau protein by an unconventional mechanism

    Neurobiol Dis

    (2012)
  • A. Gomez-Ramos et al.

    Extracellular tau promotes intracellular calcium increase through M1 and M3 muscarinic receptors in neuronal cells

    Mol Cell Neurosci

    (2008)
  • M. Diaz-Hernandez et al.

    Tissue-nonspecific alkaline phosphatase promotes the neurotoxicity effect of extracellular tau

    J Biol Chem

    (2010)
  • D. Simón et al.

    Proteostasis of tau. Tau overexpression results in its secretion via membrane vesicles

    FEBS Lett

    (2012)
  • S. Saman et al.

    Exosome-associated tau is secreted in tauopathy models and is selectively phosphorylated in cerebrospinal fluid in early Alzheimer disease

    J Biol Chem

    (2012)
  • D.N. Drechsel et al.

    Modulation of the dynamic instability of tubulin assembly by the microtubule-associated protein tau

    Mol Biol Cell

    (1992)
  • P. LoPresti et al.

    Functional implications for the microtubule-associated protein tau: Localization in oligodendrocytes

    Proc Natl Acad Sci USA

    (1995)
  • A. Ebneth et al.

    Overexpression of tau protein inhibits kinesin-dependent trafficking of vesicles, mitochondria, and endoplasmic reticulum: Implications for Alzheimer's disease

    J Cell Biol

    (1998)
  • M. Goedert et al.

    Cloning and sequencing of the cDNA encoding an isoform of microtubule-associated protein tau containing four tandem repeats: differential expression of tau protein mRNAs in human brain

    EMBO J

    (1989)
  • M. Goedert et al.

    Expression of separate isoforms of human tau protein: correlation with the tau pattern in brain and effects on tubulin polymerization

    EMBO J

    (1990)
  • L.I. Binder et al.

    The distribution of tau in the mammalian central nervous system

    J Cell Biol

    (1985)
  • I. Grundke-Iqbal et al.

    Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology

    Proc Natl Acad Sci USA

    (1986)
  • V.M. Lee et al.

    Neurodegenerative disorders

    Annu Rev Neurosci

    (2001)
  • F. Hernandez et al.

    Tauopathies

    Cell Mol Life Sci

    (2007)
  • A.C. Ludolph et al.

    Tauopaties with parkinsonism clinical spectrum: neuropathological basis, biological markers, and treatment options

    Eur J Neurol

    (2009)
  • A.C. Alonso et al.

    Role of abnormally phosphorylated tau in the breakdown of microtubules in Alzheimer disease

    Proc Natl Acad Sci USA

    (1994)
  • A.C. Alonso et al.

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

    Nat Med

    (1996)
  • Z. Berger et al.

    Accumulation of pathological tau species and memory loss in a conditional model of tauopathy

    J Neurosci

    (2007)
  • K. Iqbal et al.

    Mechanisms of tau-induced neurodegeneration

    Acta Neuropathol

    (2009)
  • P.V. Arriagada et al.

    Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer's disease

    Neurology

    (1992)
  • M. Rapoport et al.

    Tau is essential to β-amyloid-induced neurotoxicity

    Proc Natl Acad Sci USA

    (2002)
  • E.D. Roberson et al.

    Reducing endogenous tau ameliorates amyloid β-induced deficits in an Alzheimer's disease mouse model

    Science

    (2007)
  • J. Marx

    A new take on tau

    Science

    (2007)
  • M. Medina

    Recent developments in tau-based therapeutics for neurodegenerative diseases

    Recent Pat CNS Drug Discov

    (2011)
  • T. Jaworski et al.

    Modeling of tau-mediated synaptic and neuronal degeneration in Alzheimer's disease

    Int J Alzheimers Dis

    (2010)
  • C. Haass et al.

    Protein oligomers in neurodegeneration: lessons from the Alzheimer's amyloid beta-peptide

    Nat Rev Mol Cell Biol

    (2007)
  • K. Iqbal et al.

    Developing pharmacological therapies for Alzheimer disease

    Cell Mol Life Sci

    (2007)
  • Cited by (104)

    • Pseudoginsenoside-F11 ameliorates okadiac acid-induced learning and memory impairment in rats via modulating protein phosphatase 2A

      2021, Mechanisms of Ageing and Development
      Citation Excerpt :

      It is well-known that the main pathological features of Alzheimer’s disease (AD), one of the neurodegenerative diseases manifested as progressive memory loss and cognitive impairments (Wirths et al., 2016), are characterized by Aβ deposition, tau protein hyperphosphorylation, neuron loss and gliosis (Wirths et al., 2016). Emerging evidence reveals that tau hyperphosphorylation plays a central role in the pathogenesis of AD (Giacobini and Gold, 2013; Medina and Avila, 2014; Simic et al., 2016; Keshav and Marwan, 2015). It is reported that hyperphosphorylated tau proteins can cause synaptic transmission disorders (Zhou et al., 2017), gliacyte over-activation and neuroinflammation (Kamat et al., 2013), and eventually lead to neuronal death and cognitive impairment (Simic et al., 2016).

    • Complex roles for reactive astrocytes in the triple transgenic mouse model of Alzheimer disease

      2020, Neurobiology of Aging
      Citation Excerpt :

      These peptides then oligomerize and form amyloid plaques in the brain of AD patients and can cause synaptic dysfunction, neurodegeneration, and cognitive impairment (Hardy and Higgins, 1992). The amyloid hypothesis was later refined to include tau hyper-phosphorylation as a key pathogenic element (Kametani and Hasegawa, 2018; Medina and Avila, 2014). Hyper-phosphorylated tau proteins can also oligomerize and form neurofibrillary tangles that inhibit microtubule assembly and alter neuronal functions.

    View all citing articles on Scopus

    Chemical compounds studied in this article: Tideglusib (PubChem CIP: 11313622); Davunetide (PubChem CIP: 9832404); Lithium chloride (PubChem CIP: 433294); Lithium carbonate (PubChem CIP: 11125); Methylene blue (PubChem CIP: 6099); Leucomethylene blue (PubChem CIP: 164695); Sodium selenate (PubChem CIP: 25960); S-AMPA (PubChem CIP: 158397); Divalproex (PubChem CIP: 23663956).

    View full text