Elsevier

Neurobiology of Disease

Volume 76, April 2015, Pages 112-125
Neurobiology of Disease

α-Synuclein-mediated inhibition of ATF6 processing into COPII vesicles disrupts UPR signaling in Parkinson's disease

https://doi.org/10.1016/j.nbd.2015.02.005Get rights and content

Highlights

  • α-Synuclein inhibits efficient signaling by the unfolded protein response (UPR).

  • α-Synuclein reduces ATF6 signaling through direct physical association.

  • α-Synuclein inhibits ATF6 incorporation into COPII vesicles

  • Aberrant UPR signaling is a salient feature of Parkinson's disease (PD).

  • Study provides a novel cellular mechanism of how ER stress and the UPR contribute to neurodegeneration in PD.

Abstract

The unfolded protein response (UPR) monitors the folding environment within the endoplasmic reticulum (ER). Accumulation of misfolded proteins within the ER activates the UPR resulting in the execution of adaptive or non-adaptive signaling pathways. α-Synuclein (α-syn) whose accumulation and aggregation define the pathobiology of Parkinson's disease (PD) has been shown to inhibit ER–Golgi transit of COPII vesicles. ATF6, a protective branch of the UPR, is processed via COPII mediated ER–Golgi transit following its activation via ER stress. Using cellular PD models together with biochemical reconstitution assays, we showed that α-syn inhibited processing of ATF6 directly through physical interactions and indirectly through restricted incorporation into COPII vesicles. Impaired ATF6 signaling was accompanied by decreased ER-associated degradation (ERAD) function and increased pro-apoptotic signaling. The mechanism by which α-syn inhibits ATF6 signaling expands our understanding of the role ER stress and the UPR play in neurodegenerative diseases such as PD.

Introduction

Parkinson's disease (PD) is characterized by the abnormal accumulation and aggregation of α-synuclein, which leads to the selective loss of dopaminergic neurons (DA-neurons) (Braak et al., 2003). Accumulation of α-synuclein (α-synWT) in idiopathic PD or expression of the missense point mutants (E46K, A30P, A53T) in familial PD is strongly associated with age of disease onset and severity (Narhi et al., 1999). The mechanism of how α-syn disturbs cellular function leading to PD pathology (e.g., loss of DA-neurons, Lewy bodies) remains a topic of ongoing investigation. α-Syn has been shown to localize to and accumulate within various cellular compartments and organelles such as the nucleus, mitochondria, and ER. This pattern of widespread localization by α-syn is surprising given the protein's lack of a nuclear localization signal, mitochondrial import signal, signal sequence, or an ER-retention signal such as KDEL. (Devi et al., 2008, Maroteaux et al., 1988). Of particular importance to this present study are recent findings examining human PD tissue and PD animal models, which showed α-syn accumulation within the ER lumen (Colla et al., 2012a, Colla et al., 2012b, Oaks et al., 2013).

Previous studies have demonstrated that α-syn over-expression strongly inhibited ER–Golgi transport of COPII vesicles, thereby disrupting normal transit and processing of proteins within the secretory pathway. α-Syn-mediated inhibition on secretory function has been verified using a variety of secretory substrates and reporters such as the human Dopamine Transporter (hDAT) and the GFP-tagged viral glycoprotein, VSVG (Oaks et al., 2013, Thayanidhi et al., 2010). Several studies have found that α-synWT or the A53T (α-synA53T) variant (herein collectively referred to as α-syn) inhibited SNARE and RAB function leading to declines in ER–Golgi transit of COPII vesicle while over-expression of several secretory accessory proteins (Ykt6, Ypt1p, Sec22b, RAB1, RAB3a, and RAB8A) reduced α-syn dependent toxicities in cellular PD models (Cooper et al., 2006, Gitler et al., 2008, Sancenon et al., 2012, Thayanidhi et al., 2010). Chemical screens have led to the discovery of small molecules, which increased ER–Golgi transport, thereby decreasing cytotoxicity associated with α-syn over-expression (Su et al., 2011). These studies collectively support the detrimental effects that α-syn has on proper secretory function and provide motivation for the discovery and biochemical elucidation of how such effects may be causal in the premature cell death of α-syn over-expressing neurons such as those found in the SN of PD patients.

The unfolded protein response (UPR) serves as a sentinel of protein folding within the ER, maintaining both the quantity and quality of protein folding (Ron and Walter, 2011). Upon activation, the UPR orchestrates cellular changes in transcription, translation, and protein degradation. These changes are mediated through the three canonical sensors of the metazoan UPR; IRE1, ATF6, and PERK. Each sensor detects misfolded proteins via their ER luminal domains and signals through their respective cytoplasmic domains. Upon activation, full-length ATF6 (ATF6FL) transits to the Golgi via COPII vesicular transport, where sequential cleavage by regulated intramembrane proteolysis (RIP) occurs, releasing the soluble cytosolic domain (aa1-373), a basic leucine zipper (bZip) transcription factor (ATF6N) (Haze et al., 1999). Activated IRE1 proceeds to cleave the bound unspliced XBP1 mRNA (XBP1U), to produce the mature transcription factor, XBP1S (Yoshida et al., 2001). Cooperation between the ATF6 and IRE1-XBP1 signaling pathways occurs through the heterodimerization of the transcriptional domains inducing strong expression of genes necessary for maintaining ER folding homeostasis, including ER resident chaperones, ER associated degradation [ERAD] genes, and genes involved in regulation of the secretory pathway (Yamamoto et al., 2007, Yoshida et al., 2003). Increased induction of UPR-dependent genes is one component of the Integrated Stress Response (ISR), during which the individual branches of the UPR work in concert to promote cellular recovery and survival (Ron and Walter, 2007).

While there is accumulating evidence supporting involvement of the UPR in the pathobiology of PD, it remains unclear if UPR activation and signaling are protective, benign, or causative in PD (Bellucci et al., 2011, Gorbatyuk et al., 2012, Hoozemans and Scheper, 2012, Hoozemans et al., 2007, Nijholt et al., 2012, Valdés et al., 2014). Moreover, interactions between α-syn and components of the UPR remain unexplored. Analysis of PD tissue and PD models has focused mainly on observations of the PERK branch, with results showing increased expression of the PERK downstream targets, p-eIF2α, CHOP, and ATF4 (Bellucci et al., 2011, Colla et al., 2012a, Colla et al., 2012b, Gorbatyuk et al., 2012, Holtz and O'Malley, 2003, Hoozemans and Scheper, 2012, Hoozemans et al., 2007, Mutez et al., 2014, Nijholt et al., 2012). As previously mentioned, several studies have shown that α-syn inhibits COPII ER–Golgi transport, a pathway necessary for activation and signaling by ATF6. α-Syn has also been shown to form aggregates within the ER lumen suggesting that cytotoxicity may occur through a combination of α-syn localization to the ER and cytoplasm (Colla et al., 2012a, Colla et al., 2012b, Oaks et al., 2013). Other studies have provided evidence supporting the protective role of ATF6 in preventing degeneration of DA-neurons in chemically induced PD models and in the protection of cells from chronic ER stress (Egawa et al., 2011, Hashida et al., 2012, Wu et al., 2007). Therefore, diminished ATF6-processing through the effects of ER and cytoplasmic localized α-syn inhibition on COPII ER–Golgi transport would ultimately disrupt signal integration among the UPR branches through reduced cooperation between ATF6N and XBP1S. Collectively, these and other studies provide compelling evidence that accumulation of α-syn would have deleterious effects on the ability of the UPR to induce and maintain proper folding programs within the ER lumen while inhibiting the ability of the ER to efficiently remove misfolded proteins through ER-quality control measures such as ERAD.

In this report we demonstrate the ability of α-syn to disrupt UPR signaling through a combination of ER luminal and cytoplasmic insults. First, we show that GRP78, an ER resident chaperone, and general marker of ER stress levels is increased in DA-neurons of the SN from PD patient samples when compared to age-matched control tissue. Increased expression of GRP78 in PD is contradicted by the accompanying observation of decreased expression levels of ATF6FL and decreased nuclear localization of the cytosolic signaling domain, ATF6N. Second, we demonstrated that α-syn over-expression results in a decay of downstream UPR signaling pathways such as ERAD. Third, biophysical studies showed that α-syn formed a stable detergent-resistant complex with ATF6FL. Lastly, biochemical reconstitution of COPII budding showed that transient over-expression of α-syn reduced ATF6FL incorporation into COPII vesicles and caused deviations from the normal biophysical traits of such vesicles with observed increases in vesicle sizes and alterations to vesicular morphology. These data support the conclusions that α-syn over-expression and accumulation results in inhibition of ATF6 processing during periods of ER stress thus preventing execution of adaptive signaling programs, with the affected cell becoming increasingly incapable of adapting to ER stress, resulting in the premature death of selective neuronal populations.

Section snippets

Cell culture and transfection

The culturing and transfection of SH-SY5Y cells stably expressing α-SynWT, α-SynA53T, or vector alone and the CHO-ATF6-3XFlag stable line are described previously (Mazzulli et al., 2006, Oaks et al., 2013, Schindler and Schekman, 2009, Wersinger et al., 2003). See supplemental information for expanded protocol.

Cell viability assays

300 μl of media was recovered from 1.5 ml of media plus cells and used in the LDH release assay (Clontect Inc. — Mountain View, CA) according to the manufacturer's protocol.

qRT-PCR

qRT-PCR was

Increased ER stress is associated with decreased ATF6 activation in neurons of Parkinson's disease patient

Previous studies have linked α-syn over-expression with increased ER stress in cellular and animal models of PD (Bellucci et al., 2011, Colla et al., 2012a, Colla et al., 2012b, Holtz and O'Malley, 2003). To quantify ER stress levels and α-syn expression levels, GRP78 and α-syn was measured through semi-quantitative histological analysis comparing human age-matched Substantia nigra tissue from non-diseased (CT) and PD patients (Fig. S2E). As expected, α-syn expression levels were higher in

α-Syn compromises UPR signaling during ER stress

We have provided a biochemical mechanism, addressing why in PD and perhaps other neurodegenerative conditions, there is a failure of the UPR to return folding homeostasis back to the ER lumen, resulting in ER-dependent cell death. The data presented here supports a model in which α-syn disrupts UPR signaling directly through interactions with ATF6 and indirectly through disruption of ATF6 incorporation into COPII vesicles (Fig. 8A). α-Syn toxicity may occur through its localization to the

Author contributions

Conceived and designed the experiments: JJC, AWO, AS.

Performed the experiments: JJC, MD, EP, DB, VL, JW.

Analyzed the data: JJC, PAF, AS.

Contributed reagents/materials/analytical tools: JJC, PAF, AWO, MD, EP, DB, AS.

Wrote the manuscript: JJC, AS.

Competing financial interest or conflicts of interest

None.

Acknowledgments

This work was supported primarily by NIH grants to Anita Sidhu from the National Institute of Mental Health (RO1MH075020), the National Institute of Aging (RO1AG28108), and the National Institute of neurological Disorders and Stroke (RO1NS060041). Additional grant support to Joel J Credle came from the Parkinson's Disease Foundation (PDF.org). The Lombardi Comprehensive Cancer Center Microscopy and Imaging Shared Resource (LCCC-MISR) were funded in part by a grant from the NIH National Cancer

References (77)

  • J.J. Hoozemans et al.

    Endoplasmic reticulum: the unfolded protein response is tangled in neurodegeneration

    Int. J. Biochem. Cell Biol.

    (2012)
  • J.J. Hoozemans et al.

    Activation of the unfolded protein response in Parkinson's disease

    Biochem. Biophys. Res. Commun.

    (2007)
  • B. Kleizen et al.

    Folding of CFTR is predominantly cotranslational

    Mol. Cell

    (2005)
  • H.A. Lashuel

    Alpha-synuclein, especially the Parkinson's disease-associated mutants, forms pore-like annular and tubular protofibrils

    J. Mol. Biol.

    (2002)
  • E. Mutez

    Involvement of the immune system, endocytosis and EIF2 signaling in both genetically determined and sporadic forms of Parkinson's disease

    Neurobiol. Dis.

    (2014)
  • L. Narhi

    Both familial Parkinson's disease mutations accelerate alpha-synuclein aggregation

    J. Biol. Chem.

    (1999)
  • A.W. Oaks et al.

    Synuclein modulation of monoamine transporters

    FEBS Lett.

    (2011)
  • L. Orci et al.

    A new type of coated vesicular carrier that appears not to contain clathrin: its possible role in protein transport within the Golgi stack

    Cell

    (1986)
  • J. Varkey

    Membrane curvature induction and tubulation are common features of synucleins and apolipoproteins

    J. Biol. Chem.

    (2010)
  • Y. Wang et al.

    Activation of ATF6 and an ATF6 DNA binding site by the endoplasmic reticulum stress response

    J. Biol. Chem.

    (2000)
  • J. Wu

    ATF6alpha optimizes long-term endoplasmic reticulum function to protect cells from chronic stress

    Dev. Cell

    (2007)
  • K. Yamamoto

    Transcriptional induction of mammalian ER quality control proteins is mediated by single or combined action of ATF6alpha and XBP1

    Dev. Cell

    (2007)
  • H. Yoshida et al.

    XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor

    Cell

    (2001)
  • H. Yoshida et al.

    A time-dependent phase shift in the mammalian unfolded protein response

    Dev. Cell

    (2003)
  • J.F. Abisambra

    Tau accumulation activates the unfolded protein response by impairing endoplasmic reticulum-associated degradation

    J. Neurosci.

    (2013)
  • A. Bellucci

    Induction of the unfolded protein response by α-synuclein in experimental models of Parkinson's disease

    J. Neurochem.

    (2011)
  • J.L. Biedler et al.

    Multiple neurotransmitter synthesis by human neuroblastoma cell lines and clones

    Cancer Res.

    (1978)
  • C.Y. Chung

    Identification and rescue of α-synuclein toxicity in Parkinson patient-derived neurons

    Science

    (2013)
  • E. Colla et al.

    Accumulation of toxic α-synuclein oligomer within endoplasmic reticulum occurs in α-synucleinopathy in vivo

    J. Neurosci.

    (2012)
  • E. Colla

    Endoplasmic reticulum stress is important for the manifestations of α-synucleinopathy in vivo

    J. Neurosci.

    (2012)
  • A.A. Cooper

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

    Science

    (2006)
  • V.H. Cornejo et al.

    The unfolded protein response in Alzheimer's disease

    Semin. Immunopathol.

    (2013)
  • J. Diao et al.

    Native α-synuclein induces clustering of synaptic-vesicle mimics via binding to phospholipids and synaptobrevin-2/VAMP2

    Elife

    (2013)
  • P.E. Duffy et al.

    Phase and electron microscopic observations of Lewy bodies and melanin granules in substantia nigra and locus caeruleus in Parkinsons disease

    J. Neuropathol. Exp. Neurol.

    (1965)
  • D.L. Eaton et al.

    Concise review of the glutathione S-transferases and their significance to toxicology'

    Toxicol. Sci.

    (1999)
  • P.A. Garris et al.

    Efflux of dopamine from the synaptic cleft in the nucleus accumbens of rat brain

    J. Neurosci.

    (1994)
  • A.D. Gitler

    The Parkinson's disease protein alpha-synuclein disrupts cellular Rab homeostasis

    Proc. Natl. Acad. Sci. U. S. A.

    (2008)
  • K. Hashida

    ATF6alpha promotes astroglial activation and neuronal survival in a chronic mouse model of Parkinson's disease

    PLoS One

    (2012)
  • Cited by (83)

    • Proteostasis in Parkinson's disease: Recent development and possible implication in diagnosis and therapeutics

      2023, Ageing Research Reviews
      Citation Excerpt :

      Intraventricular PR001 treatment elevated GCase expression, increased GCase enzyme activity, decreased the buildup of GCase glycolipid substrates, corrected neuroinflammation, and improved behavioral outcomes (Abeliovich et al., 2021). Clinical trials have been registered for testing GLP-1 receptor agonists as they have been found to inhibit glycogen synthase kinase 3B which in turn inhibits phosphorylation of α-syn and tau such as Exenatide, Lixisenatide, Liraglutide, Semaglutide and NLY01 (NCT04154072) however, their clinical use is yet to be validated (Credle et al., 2015a, 2015b). Non-receptor tyrosine kinase cAbl (Abelson tyrosine kinase) activation can initiate α- synucleinopathy by inducing oxidative stress, accumulation of α-syn, and phosphorylated Ser129 α-syn and affects E3 ubiquitin ligase activity of PARKIN, causing neurodegeneration of dopaminergic neurons.

    • Dual Role of Inositol-requiring Enzyme 1α–X-box Binding protein 1 Signaling in Neurodegenerative Diseases

      2022, Neuroscience
      Citation Excerpt :

      Notably, pathological protein aggregates are typically found in the cytoplasm and not in the ER in most neurodegenerative disorders. Cytoplasmic protein aggregates are thought to trigger ER stress through multiple pathways, including the disruption of the normal protein folding process, impaired ER-associated degradation (ERAD) function, and ER calcium overload (Zampese et al., 2011; Usenovic et al., 2012; Abisambra et al., 2013; Credle et al., 2015). To alleviate ER stress, cells activate a compensatory mechanism called the unfolded protein response (UPR) that reduces protein load (Schwarz and Blower, 2016).

    • Rescue of α-synuclein aggregation in Parkinson's patient neurons by synergistic enhancement of ER proteostasis and protein trafficking

      2022, Neuron
      Citation Excerpt :

      UPR activation has been documented in various synucleinopathy models including α-syn-overexpressing yeast (Cooper et al., 2006), A53T transgenic mice (Colla et al., 2012a, 2018), and induced pluripotent stem cell (iPSC)-derived cortical neuron models (Chung et al., 2013; Heman-Ackah et al., 2017). Evidence of UPR activation has been shown in the substantia nigra of post-mortem PD brains (Credle et al., 2015; Heman-Ackah et al., 2017; Hoozemans et al., 2007). Overexpression of the ER chaperone GRP78 can also reduce neurodegeneration in α-syn-expressing animal models (Gorbatyuk et al., 2012), further emphasizing the importance of maintaining ER proteostasis in neuronal health.

    View all citing articles on Scopus
    View full text