Review
Cell systems and the toxic mechanism(s) of α-synuclein

https://doi.org/10.1016/j.expneurol.2007.05.022Get rights and content

Abstract

Mutations in the SNCA gene are causal for familial Parkinson disease/Lewy body disease. α-Synuclein is a small acidic protein that binds loosely to the surface of vesicles and may play a role in synaptic dynamics, although its normal function remains somewhat unclear. What is clear is that point mutations or increased expression of wild type α-synuclein causes disease. A great deal of literature supports the overall hypothesis that α-synuclein is damaging to neurons because it is inherently prone to aggregation; mutations or increased concentration of the protein both increase this tendency. An unproven, but popular, contention is that the toxic species are small oligomers that are relatively soluble, which may react with membranes to damage key processes within the cell. The details of this process, especially in determining the order of events and the requirement of particular processes in cell death, are unclear. Derangements in vesicle processing, including synaptic function, protein turnover, mitochondrial function and oxidative stress, have all been suggested to occur. Whether there is a sequence of events or whether these are interacting effects is unclear, but the outcome is to trigger cell death, by both apoptotic and non-apoptotic mechanisms depending on the system studied. In this article, we develop a framework for thinking about α-synuclein in terms of initiating events and secondary processes that are required to trigger neuronal dysfunction and cell death.

Introduction

Without α-synuclein, understanding the relationships between familial and sporadic forms of Parkinson disease (PD) would be extremely difficult. Mutations in the SNCA gene produce a range of phenotypes from PD to diffuse Lewy body disease (DLBD), the link being that α-synuclein is deposited in the pathological hallmark of PD/DLBD, the Lewy body. Quantitative mutations, such as triplication of the SNCA locus, lead to increased expression of the gene and to a disease with PD/DLBD features (Singleton et al., 2003). All mutations are dominant, suggesting that there is a gain of function and that the point mutations and multiplications have similar mechanisms. This leads to the argument that α-synuclein can be considered a causative agent for PD and related disorders (Cookson, 2005).

These considerations leave us with the simplest possible sketch of the pathogenesis of PD: that α-synuclein is an initiator of damage and the final outputs, after many years, are cell loss and Lewy body formation. Formally, α-synuclein tells us about etiology in familial disease and is likely to be close to the etiology of sporadic disease. We also know the range of disease presentations associated with the etiology. This is often represented diagrammatically with α-synuclein by an arrow pointing at PD, occasionally with a question mark over the arrow to indicate ignorance. The obvious limitation with this level of analysis is that it sidesteps the question of why α-synuclein causes cell loss. Why synuclein is toxic might be best broken down into two sets of questions. First, what are the immediate biochemical effects of mutations or increased expression of α-synuclein that might be associated with damaging effects? Although not certain, the prevailing thought is that protein aggregation is important in pathogenesis, similar to other ‘toxic proteins’ associated with a number of neurodegenerative diseases (Taylor et al., 2002). Second, what are the downstream events that mediate the toxic effects of α-synuclein on the neuron? As will be discussed in this review, there are several theories of why α-synuclein might cause neuronal damage and a major challenge to the field is to elucidate those that are critical from those that are secondary effects.

Where cell-based models might be useful is in delineating the mechanisms of pathogenesis in simple ways. For example, because it is possible to look at the timing of aggregation relative to downstream events we might be able to order the events related to α-synuclein toxicity. This can be achieved using an inducible system and following toxicity over time (e.g., Tanaka et al., 2001). One might also be able to perform multiple manipulations on cells and establish which are beneficial to α-synuclein toxicity without prejudging which pathways are important, as has been done in yeast (e.g., Willingham et al., 2003). What cell-based assays are not are measures of pathogenesis in the intact organism – for these, one will have to use systems where α-synuclein is expressed in the brain, discussed elsewhere in this issue. Here we will discuss only models that use cell-based systems, including both mammalian cell cultures and yeast models. The basic supposition of all these models is that α-synuclein can be expressed at sufficient concentration to cause the cell to die or to become dysfunctional. We will discuss the model systems in the context of known or proposed mechanisms involved in cellular toxicity.

Section snippets

Protein aggregation, oligomers and pores

As discussed briefly above, a major theory regarding α-synuclein is that it is prone to form aggregates (Cookson, 2005). The aggregation process initially forms oligomeric species that are relatively soluble; these oligomers might then self-assemble into fibrillar structures that are insoluble. A major, but untested, concept is that the process of fibrillization is important to the formation of Lewy bodies as these organized structures become deposited (Mukaetova-Ladinska and McKeith, 2006).

The ER–Golgi connection

A difficulty in deciding how proteins are toxic is the identification of which effects are primary, and therefore critical, and which might be secondary. An important approach is to use relatively unbiased screens of multiple pathways, illustrated in one study using yeast cells to look for modifiers of α-synuclein toxicity (Willingham et al., 2003). The results show that suppressors of α-synuclein toxicity are not randomly distributed in different groups but could be classified by gene ontology

Out to the synapse

When discussing alterations in vesicle dynamics in yeast cells that result from expression of α-synuclein, we have to consider what the equivalent processes are in neurons. Given that the major function of neurons is to produce and release neurotransmitters packaged into vesicles, one possibility for such a process is synaptic transmission. α-Synuclein was originally named, in part, for being a synaptic protein and seems to be largely presynaptic (Maroteaux et al., 1988). Some recent

Mitochondrial function

A longstanding theory in PD research is that mitochondrial function is an important clue for the preferential cell loss seen for some groups of neurons in the brain. This has been bolstered by the observation that each of three genes that cause recessive parkinsonism are associated, directly or indirectly, with mitochondrial function (Shen and Cookson, 2004, Clark et al., 2006, Park et al., 2006). Although recessive parkinsonism is a very different phenotype from Lewy body disease, the common

Derangements of protein turnover

There are two major systems for protein degradation in mammalian cells, the ubiquitin–proteasome system (UPS) and lysosomal systems including autophagy (reviewed in Rubinsztein, 2006). Several studies in cell culture have implicated an effect of α-synuclein on both of these.

Inhibition of UPS function may be a general mechanism for several proteins associated with disease that are prone to misfolding. Bence et al. elegantly demonstrated this a few years ago, showing that net proteasome function

Oxidative stress and dopamine

Many of the concepts discussed above are common to multiple neurodegenerative diseases. Another commonly discussed mechanism for neuronal damage is oxidative stress, which is often linked specifically to PD because dopamine (DA) is highly prone to undergo oxidation. Additionally, the metabolism of DA produces toxic metabolites (Stokes et al., 1999) and can generate reactive oxygen species (ROS) (Maguire-Zeiss et al., 2005). There is some evidence for a direct connection of DA oxidation to

Triggering cell death: is there a role for apoptosis?

Aggregated α-synuclein is unambiguously toxic to neurons, though the means by which this causes cellular death is as unclear as the mechanisms that trigger the initial aggregation. In cellular models, increased WT α-synuclein has been shown to correlate with apoptotic markers in neurons (Saha et al., 2000) glia (Stefanova et al., 2001) and lymphoblasts (Kim et al., 2004). Classic markers such as chromatin condensation, nuclear and DNA fragmentation and cytochrome C release may also be observed

Summary

Despite there being a great deal of work on how α-synuclein damages neurons, only a few clear principles have been established. In this article, we have followed the following framework for understanding why α-synuclein is toxic. First, it seems likely that there are some very proximal events that initiate toxicity, which relate to the unusual biochemistry of the α-synuclein protein. The prevailing hypothesis is that aggregation of the protein into small soluble oligomeric species is likely to

Acknowledgment

This research was supported by the Intramural Research Program of the NIH, National Institute on Aging.

References (74)

  • E. Lindersson et al.

    Proteasomal inhibition by alpha-synuclein filaments and oligomers

    J. Biol. Chem.

    (2004)
  • K.A. Maguire-Zeiss et al.

    Synuclein, dopamine and oxidative stress: co-conspirators in Parkinson's disease?

    Brain Res. Mol. Brain Res.

    (2005)
  • E.B. Mukaetova-Ladinska et al.

    Pathophysiology of synuclein aggregation in Lewy body disease

    Mech. Ageing Dev.

    (2006)
  • M. Orth et al.

    alpha-Synuclein expression in HEK293 cells enhances the mitochondrial sensitivity to rotenone

    Neurosci. Lett.

    (2003)
  • L. Petrucelli et al.

    Parkin protects against the toxicity associated with mutant alpha-synuclein: proteasome dysfunction selectively affects catecholaminergic neurons

    Neuron

    (2002)
  • M.C. Quilty et al.

    alpha-Synuclein is upregulated in neurones in response to chronic oxidative stress and is associated with neuroprotection

    Exp. Neurol.

    (2006)
  • S. Sarkar et al.

    Trehalose, a novel mTOR-independent autophagy enhancer, accelerates the clearance of mutant huntingtin and {alpha}-synuclein

    J. Biol. Chem.

    (2007)
  • R. Sharon et al.

    The formation of highly soluble oligomers of alpha-synuclein is regulated by fatty acids and enhanced in Parkinson's disease

    Neuron

    (2003)
  • J. Shen et al.

    Mitochondria and dopamine: new insights into recessive parkinsonism

    Neuron

    (2004)
  • H. Snyder et al.

    Aggregated and monomeric alpha-synuclein bind to the S6′ proteasomal protein and inhibit proteasomal function

    J. Biol. Chem.

    (2003)
  • S. Turnbull et al.

    alpha-Synuclein implicated in Parkinson's disease catalyses the formation of hydrogen peroxide in vitro

    Free Radical Biol. Med.

    (2001)
  • R. Ved et al.

    Similar patterns of mitochondrial vulnerability and rescue induced by genetic modification of alpha-synuclein, parkin, and DJ-1 in Caenorhabditis elegans

    J. Biol. Chem.

    (2005)
  • M.J. Volles et al.

    Relationships between the sequence of alpha-synuclein and its membrane affinity, fibrillization propensity, and yeast toxicity

    J. Mol. Biol.

    (2007)
  • J.L. Webb et al.

    alpha-Synuclein is degraded by both autophagy and the proteasome

    J. Biol. Chem.

    (2003)
  • C. Wersinger et al.

    Attenuation of dopamine transporter activity by alpha-synuclein

    Neurosci. Lett.

    (2003)
  • S. Xu et al.

    Oxidative stress induces nuclear translocation of C-terminus of alpha-synuclein in dopaminergic cells

    Biochem. Biophys. Res. Commun.

    (2006)
  • F. Yu et al.

    Impairment of redox state and dopamine level induced by alpha-synuclein aggregation and the prevention effect of hsp70

    Biochem. Biophys. Res. Commun.

    (2005)
  • W. Zhou et al.

    DJ-1 up-regulates glutathione synthesis during oxidative stress and inhibits A53T alpha-synuclein toxicity

    J. Biol. Chem.

    (2005)
  • M. Zhu et al.

    The association of alpha-synuclein with membranes affects bilayer structure, stability, and fibril formation

    J. Biol. Chem.

    (2003)
  • M.J. Baptista et al.

    Co-ordinate transcriptional regulation of dopamine synthesis genes by alpha-synuclein in human neuroblastoma cell lines

    J. Neurochem.

    (2003)
  • N.F. Bence et al.

    Impairment of the ubiquitin–proteasome system by protein aggregation

    Science

    (2001)
  • D.E. Cabin et al.

    Synaptic vesicle depletion correlates with attenuated synaptic responses to prolonged repetitive stimulation in mice lacking alpha-synuclein

    J. Neurosci.

    (2002)
  • R. Cappai et al.

    Dopamine promotes alpha-synuclein aggregation into SDS-resistant soluble oligomers via a distinct folding pathway

    FASEB J.

    (2005)
  • I.E. Clark et al.

    Drosophila pink1 is required for mitochondrial function and interacts genetically with parkin

    Nature

    (2006)
  • M.R. Cookson

    The biochemistry of Parkinson's disease

    Annu. Rev. Biochem.

    (2005)
  • A.A. Cooper et al.

    alpha-Synuclein blocks ER–Golgi traffic and Rab1 rescues neuron loss in Parkinson's models

    Science

    (2006)
  • A.M. Cuervo et al.

    Impaired degradation of mutant alpha-synuclein by chaperone-mediated autophagy

    Science

    (2004)
  • Cited by (162)

    • Neuroprotective effects of pulicaria undulata essential oil in rotenone model of parkinson's disease in rats: Insights into its anti-inflammatory and anti-oxidant effects

      2020, South African Journal of Botany
      Citation Excerpt :

      One of the characteristic pathological hallmarks of PD is the occurrence of intracellular protein aggregates known as LBs (Ferrer, 2009). α-Synuclein comprises the major fraction of LBs (Cookson and van der Brug, 2008). Treatment with rotenone is known to induce several modifications in the α-synuclein protein (Mirzaei et al., 2006) involving up-regulation of its expression (Betarbet et al., 2000).

    View all citing articles on Scopus
    View full text