Elsevier

Experimental Neurology

Volume 225, Issue 1, September 2010, Pages 210-218
Experimental Neurology

Elevated tauopathy and alpha-synuclein pathology in postmortem Parkinson's disease brains with and without dementia

https://doi.org/10.1016/j.expneurol.2010.06.017Get rights and content

Abstract

Parkinson's disease (PD), a progressive neurodegenerative disease, results in abnormal accumulation of insoluble alpha-synuclein (α-Syn) in dopaminergic neurons. Here we examined tauopathic changes and the α-Syn/p-GSK-3β/proteasome pathway in postmortem striata and inferior frontal gyri (IFG) from patients with PD and PD with dementia (PDD). In both PD and PDD, α-Syn levels were high, especially the insoluble form of this protein; in PDD, insoluble α-Syn levels were persistently higher than PD across both brain regions. Levels of p-GSK-3β phosphorylated at Tyr 216, which hyperphosphorylates Tau to produce toxic pathological forms of p-Tau, were higher in striata of both PD and PDD compared to controls, but were unaltered in IFG. While proteasomal activity was unchanged in striatum of PD and PDD, such activity was diminished in the IFG of both PD and PDD. A decrease in 19S subunit of the proteasomes was seen in IFG of PDD, while lower levels of 20S subunits were seen in striatum and IFG of both PD and PDD patients. Parkin levels were similar in PD and PDD, suggesting lack of involvement of this protein. Most interestingly, tauopathic changes were noted only in striatum of PD and PDD, with increased hyperphosphorylation seen at Ser262 and Ser396/404; increases in Ser202 levels were seen only in PD but not in PDD striatum. We were unable to detect any tauopathy in IFG in either PD or PDD despite increased levels of α-Syn, and decreased proteasomal activity, and is probably due to lack of increase in p-GSK-3β in IFG. Unlike Alzheimer's disease where tauopathy is more globally observed in diverse brain regions, our data demonstrates restricted expression of tauopathy in brains of PD and PDD, probably limited to dopaminergic neurons of the nigrostriatal region.

Introduction

α-Synuclein (α-Syn) is ubiquitously expressed in brain and is highly enriched in presynaptic nerve terminals, where its chief physiological function is the regulation of synaptic levels of monoamine neurotransmitters through modulation of vesicular release (Murphy et al., 2000) and their cognate transporters (Wersinger and Sidhu, 2005, Wersinger et al., 2006a, Wersinger et al., 2006b). Overexpression of α-Syn, through its gene duplication and triplication, is linked to idiopathic Parkinson's disease (PD), while its A30P and A53T mutants cause the autosomal dominant forms of familial PD (Hofer et al., 2004, Krüger et al., 1998, Polymeropoulos et al., 1997). In pathological states, α-Syn becomes misfolded, aggregates and accumulates in neuronal inclusion bodies, Lewy bodies (LBs), seen in PD and other synucleinopathies (Goedert and Spillantini, 1998, Hofer et al., 2004). Tau, a microtubule binding protein, is most commonly linked to Alzheimer's disease (AD) and other tauopathies, where, after hyperphosphorylation, it accumulates in neurons as neurofibrillary tangles [NFTs] (Joachim et al., 1987).

Despite differences in clinical features, pathological and experimental evidence increasingly indicates that there is considerable overlap between tauopathies and synucleinopathies, reinforcing the notion that these diseases may be mechanistically linked. Thus, in AD, 50–60% of patients with NFTs have α-Syn-containing LBs (Lippa et al., 1998, Iseki et al., 1999, Arai et al., 2001, Szpak et al., 2001, Burns et al., 2005, Griffin et al., 2006) and in AD patients with clinically detected extrapyramidal signs, 50% of the patients showed extensive α-Syn pathology colocalized in the substantia nigra with p-Tau (Mori et al., 2002). In PD and in dementia with LBs, costaining of p-Tau has been observed (Duda et al., 2002, Yamaguchi et al., 2005) in 30–40% of the LBs in the nucleus basalis of Meynert and locus coerulus and in 10–30% of LBs in the medulla (Yamaguchi et al., 2005). Extensive overlap in α-Syn and p-Tau pathology has been noted in patients with the A53T mutation (Yamazaki et al., 2000, Kotzbauer et al., 2004), in the Parkinsonism–Dementia complex of Guam (Forman et al., 2002), in dementia with LBs (Yancopoulou et al., 2005), and in familial frontotemporal dementia and progressive aphasia (Hishikawa et al., 2003).

Despite this wealth of pathological information, the molecular and cellular interplay between α-Syn and p-Tau leading to their pathological codeposition is not understood. Under normal physiological conditions, α-Syn is highly soluble. Under pathological conditions, which include oxidative stress and overexpression, the protein becomes insoluble, self-aggregates, and accumulates into LBs (Nemes et al., 2004, Lippa et al., 1998, Iseki et al., 1999, Mori et al., 2002). Similar to α-Syn, Tau is a highly soluble protein that becomes insoluble by pathological hyperphosphorylation at specific sites with ensuing conformational changes and accumulation of the protein into NFTs. Molecular evidence suggests a direct interaction between these proteins, and when incubated together in vitro, α-Syn serves as a seed to accelerate the aggregation of Tau (Kotzbauer et al.,2004).

Using the MPTP mouse neurotoxin model of PD, as well as MPP+ cellular models of PD, we recently demonstrated that increases in α-Syn can initiate and sustain Tau hyperphosphorylation both in vivo and in vitro, with coprecipitation of these proteins (Duka et al., 2006, Duka and Sidhu, 2006, Kozikowski et al., 2006, Duka et al., 2009). The hyperphosphorylation of Tau was absolutely dependent on the presence of α-Syn, as indicated by lack of any p-Tau formation in MPTP-treated α-Syn−/− mice or in neuronal cells lacking α-Syn (Duka et al., 2006, Duka et al., 2009). Moreover, we found that upon oxidative stress by MPTP or MPP+, α-Syn induced p-Tau formation through specific activation and recruitment of p-GSK-3β, activated by autophosphorylation at Tyr216 (Kozikowski et al., 2006, Duka et al., 2009). GSK-3β is a proline-directed serine/threonine kinase, ubiquitously expressed in mammalian tissues, and epitopes phosphorylated by GSK-3β are among the pathological phosphorylation sites of Tau seen in NFTs and paired helical filaments (PHFs) of Alzheimer's disease, and GSK-3β is a major kinase implicated in Tau hyperphosphorylation (Baum et al., 1995).

To examine the clinical relevance of our previous findings, the current studies were undertaken, and these are the first neurochemical examination of the α-Syn/p-Tau/GSK-3β pathway in human postmortem striata and IFG from PD and PD with dementia (PDD). The results show higher neurodegeneration in PDD compared to PD, with tauopathy restricted to striata of both diseases, along with decreased proteasomal activity in the IFG of PDD. Our studies show that unlike Alzheimer's disease where tauopathy is a more global event, tauopathy in brains of PD and PDD has a restricted expression and is limited to dopaminergic neurons of the nigrostriatal region, possibly due to increased dopamine-linked oxidative stress of the latter region.

Section snippets

Materials

The antibodies used in this study are as follows: anti-DAT, MAB369 and TAU MAB361 both from Millipore (Temecula, CA); anti-α-Syn catalog no. 610787, anti-GSK-3β catalog no. 612313, and anti-pGSK-3B – Purified Mouse anti-GSK-3B (pY216) catalog no. 612313, all from BD Transduction Labs (San Jose, CA); anti-parkin ab77924, from Abcam Inc. (Cambridge, MA); and anti-tyrosine hydroxylase sc-25269 and anti-β-actin SC-1616, both from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA). The CP-13 and PHF-1

Expression levels of tyrosine hydroxylase and dopamine transporter in human post mortem brains of PD, PDD, and age-matched controls

Since decreases in tyrosine hydroxylase (TH) and dopamine transporter (DAT) levels indicate the extent of loss of monoaminergic neurons and loss of dopaminergic nerve terminals, respectively, we examined expression levels of these proteins in post mortem striata (Fig. 1A) and inferior frontal gyri (IFG; Fig. 1B) from PD, PDD, and controls using Western blots. In striata (Fig. 1A), TH levels were decreased by ~ 40% in PD and PDD cases (p < 0.01). In the IFG (Fig. 1B), TH levels were significantly

Discussion

We show here for the first time an increased state of tauopathy and an increase in associated proteins that modulate this pathway in Parkinson's disease, α-Syn and p-GSK-3β, in postmortem striata of both PD and PDD patients. Our findings demonstrate a large increase in Tau abnormally hyperphosphorylated at Ser202, Ser262, and Ser396/404 in the striatum of PD patients, with a similar increase seen in Ser262 and Ser396/404 in PDD patients; notably, Ser202 levels were not increased in striatum of

Acknowledgments

We are grateful to Peter Davies (Albert Einstein College of Medicine, NY) for the generous gift of the CP-13 and PHF-1 antibodies. We are grateful to Dr. Dianca Graham for work on unpublished studies that enabled us to form some of our conclusions. We are grateful to the families of the many patients for their generosity in donating the organs that made this study possible and to Sun Health Research Institute Brain Donation Program of Sun City, Arizona for the provision of human brain tissue.

References (42)

  • K. Schindowski et al.

    Alzheimer's disease-like tau neuropathology leads to memory deficits and loss of functional synapses in a novel mutated tau transgenic mouse without any motor deficits

    Am. J. Pathol.

    (2006)
  • C. Wersinger et al.

    Comparative analyses of alpha synuclein expression levels in rat brain tissue and transfected cells

    Neurosci. Lett.

    (2004)
  • J. Zhong et al.

    Hyperphosphorylated tau in SY5Y cells: similarities and dissimilarities to abnormally hyperphosphorylated tau from Alzheimer disease brain

    FEBS Lett.

    (1999)
  • K. Beyer et al.

    Molecular pathology of Lewy body diseases

    Int. J. Mol. Sci.

    (2009)
  • J. Burns et al.

    The pathology of the substantia nigra in Alzheimer disease with extrapyramidal signs

    Neurology

    (2005)
  • J. Duda et al.

    Concurrence of alpha-synuclein and tau brain pathology in the Contursi kindred

    Acta Neuropathol (Berl)

    (2002)
  • T. Duka et al.

    alpha-Synuclein induces hyperphosphorylation of TAU in the MPTP model of Parkinsonism

    FASEB J.

    (2006)
  • T. Duka et al.

    The neurotoxin MPP induces hyperphosphorylation of Tau in the presence of alpha-synuclein in SHSY-5Y neuroblastoma cells

    Neurotox. Res.

    (2006)
  • T. Duka et al.

    alpha-Synuclein contributes to GSK-3beta-catalyzed Tau phosphorylation in Parkinson's disease models

    FASEB J.

    (2009)
  • M. Goedert et al.

    Lewy body diseases and multiple system atrophy as alpha-synucleinopathies

    Mol. Psychiatry

    (1998)
  • W. Griffin et al.

    Interleukin-1 mediates Alzheimer and Lewy body pathologies

    J. Neuroinflammation

    (2006)
  • Cited by (167)

    • An Update on Peripheral Blood Extracellular Vesicles as Biomarkers for Parkinson's Disease Diagnosis

      2023, Neuroscience
      Citation Excerpt :

      Tau protein is a pathological marker for certain neurodegenerative diseases, including AD (Spillantini and Goedert 2013). The latest evidence disclosed an association between the tau gene (MAPT) and sporadic PD, suggesting the potential role of tau in PD (Simon-Sanchez et al. 2009; Edwards et al. 2010; Wills et al. 2010). In 2016, Shi and co-workers demonstrated that tau was detectable in blood, and they quantified exosomal tau using a kind of single molecule array kit—Simoa (Shi et al. 2016).

    View all citing articles on Scopus
    View full text