Review
Autophagy Induction as a Therapeutic Strategy for Neurodegenerative Diseases

https://doi.org/10.1016/j.jmb.2019.12.035Get rights and content

Highlights

  • Autophagy delivers cytoplasmic cargoes to lysosomes for degradation.

  • It degrades many aggregate-prone proteins responsible for neurodegenerative disease.

  • Enhancing autophagy has therapeutic potential in common neurodegenerative diseases.

  • Evidence in cells and in vivo demonstrates promising results in many disease models.

  • Outcomes may depend on how autophagy impacts disease pathogenesis.

Abstract

Autophagy is a major, conserved cellular pathway by which cells deliver cytoplasmic contents to lysosomes for degradation. Genetic studies have revealed extensive links between autophagy and neurodegenerative disease, and disruptions to autophagy may contribute to pathology in some cases. Autophagy degrades many of the toxic, aggregate-prone proteins responsible for such diseases, including mutant huntingtin (mHTT), alpha-synuclein (α-syn), tau, and others, raising the possibility that autophagy upregulation may help to reduce levels of toxic protein species, and thereby alleviate disease. This review examines autophagy induction as a potential therapy in several neurodegenerative diseases—Alzheimer’s disease, Parkinson’s disease, polyglutamine diseases, and amyotrophic lateral sclerosis (ALS). Evidence in cells and in vivo demonstrates promising results in many disease models, in which autophagy upregulation is able to reduce the levels of toxic proteins, ameliorate signs of disease, and delay disease progression. However, the effective therapeutic use of autophagy induction requires detailed knowledge of how the disease affects the autophagy-lysosome pathway, as activating autophagy when the pathway cannot go to completion (e.g., when lysosomal degradation is impaired) may instead exacerbate disease in some cases. Investigating the interactions between autophagy and disease pathogenesis is thus a critical area for further research.

Introduction

Macroautophagy (henceforth referred to as autophagy) is a major, conserved cellular process by which cells deliver cytoplasmic contents to lysosomes for degradation. This transport involves the delivery of these contents by double-membraned vesicles called autophagosomes, in contrast with other pathways, like chaperone-mediated autophagy (CMA) and microautophagy, which do not involve vesicular transport. While autophagy was initially characterized as a primordial, nonselective degradation pathway induced to counteract nutrient deprivation, it has become increasingly clear that autophagy plays a key role in the homeostasis of nonstarved cells. Critically, autophagy appears to degrade aggregate-prone proteins, damaged mitochondria, and invading pathogens, and these functions appear to be linked to a range of human diseases [1,2].

One area of particular interest is the relevance of autophagy to neurodegenerative diseases. Many neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease, manifest with the accumulation of oligomers and aggregates of misfolded proteins. As these proteins exert toxic effects on cells, lowering the levels of these proteins can be therapeutically favorable [1]. Autophagy degrades many of the toxic aggregate-prone proteins responsible for such diseases, including mutant huntingtin (mHTT), alpha-synuclein (α-syn), tau, and others [[3], [4], [5]]. Many of these proteins also cause disruptions in autophagy [6,7], raising the possibility that their ability to hinder autophagy contributes to their toxicity. In further support of a relationship between autophagy and neurodegenerative disease, some risk genes linked to neurodegenerative diseases play a role in the autophagy pathway [1,8]. This considerable body of evidence linking autophagy and neurodegeneration gives rise to the possibility that autophagy upregulation may be a viable therapeutic strategy in some neurodegenerative diseases.

Understanding the interplay between autophagy and neurodegeneration requires a knowledge of the multiple steps and regulatory pathways involved in the autophagy pathway (Fig. 1). The process of autophagy involves a series of regulated mechanical steps, including autophagosome formation, maturation, and closure [2,9]. The initial stages of autophagy are marked by cup-shaped, double-membraned phagophores, the formation of which requires PI(3)P generation by the Beclin-1-VPS34 complex [10,11]. The edges of these phagophores subsequently extend and fuse to form autophagosomes [12]. These are trafficked towards the proximity of lysosomes via the dynein machinery on microtubules [13], which allows fusion with lysosomes and degradation of autophagosomal contents. Each of these steps is subject to regulation by various upstream signaling pathways, notably via mTORC1, a major regulator of cell metabolism [14], and TFEB [15], a key transcriptional regulator of autophagy and lysosomal biogenesis. Autophagic flux, thus, depends on multiple steps and requires coordination between autophagosome biogenesis and lysosomal degradation. Interventions aimed at inducing autophagy have been targeted towards various stages in this process, potentially leading to different effects on disease progression (see Table 1).

This review will focus primarily on the potential of autophagy upregulation as a therapeutic strategy in several classes of neurodegenerative disease: Alzheimer’s disease (AD) and tauopathies, Parkinson’s disease (PD), polyglutamine diseases, and amyotrophic lateral sclerosis (ALS). Although most instances of neurodegenerative disease are sporadic, a small number of cases in each disease have been associated with disease-causing mutations in critical genes (Fig. 1). These cases have allowed for the generation of important insights into the pathogenesis of each disease and are used in many of the cell and animal models of each illness. In each section, we briefly explore how proteins linked to the relevant disease impact autophagy. Subsequently, we describe the evidence in cells and in vivo models on whether autophagy induction may be of therapeutic value. Thorough reviews have previously been published on the pathogenesis of each of the described neurodegenerative diseases [[16], [17], [18], [19]] and on the cell biology of autophagy [1,2], so these will not be explored in detail here.

Section snippets

Alzheimer’s Disease and Tauopathies

AD is a neurodegenerative disease clinically characterized by progressive dementia and cognitive impairment. The pathology of AD is defined by the presence of two main hallmark elements: intracellular accumulation of neurofibrillary tangles (formed of hyperphosphorylated tau, a microtubule-associated protein) and extracellular deposits of amyloid-β (Aβ) plaques arising from defective amyloid precursor protein (APP) processing. AD is the most common of the tauopathies, a class of

Parkinson’s Disease

Parkinson’s disease (PD) is a debilitating neurodegenerative disorder primarily characterized by progressive loss of motor control, and in many cases, cognitive decline. These symptoms are the result of the death of dopaminergic neurons in the substantia nigra pars compacta, and are associated with the accumulation of intraneuronal protein aggregates (Lewy bodies), predominantly comprised of α-synuclein (α-syn) [80]. A large body of evidence implicates defective autophagy as central to both the

Polyglutamine Diseases

There are currently nine known diseases caused by polyglutamine expansion mutations: Huntington’s disease (HD); spinal and bulbar muscular atrophy (SBMA); dentatorubropallidoluysian atrophy (DRPLA); and spinocerebellar ataxia (SCA) types 1, 2, 3, 6, 7, and 17 [144]. These mutations are encoded by the expansion of a (CAG)n trinucleotide repeat tract in the relevant proteins. For example, in HD, up to 35 CAGs are well-tolerated and considered within the range of normal variation, while 36 or more

Amyotrophic Lateral Sclerosis

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease which is characterized by progressive degeneration of upper and lower motor neurons. The majority (~90%) of ALS cases are sporadic, and only 5–10% of all cases have been reported to be familial [183]. A large number of pathogenic mutations within over 30 genes have been linked to ALS. The most commonly involved genes are superoxide dismutase (SOD1) [184], fused in sarcoma (FUS) [185], TAR DNA-binding protein 43 (TDP-43) [

Conclusion and Perspective

Deficits in protein homeostasis are a shared mechanism across neurodegenerative diseases, and therefore increasing protein clearance via autophagy is an attractive strategy that may be applicable in multiple diseases. As described above, evidence in disease models indeed demonstrates promising results in many cases, with autophagy upregulation being able to reduce the levels of toxic proteins, ameliorate signs of disease, and delay disease progression in a number of models.

Effectively utilizing

Acknowledgements

We are grateful for funding from the UK Dementia Research Institute (funded by the MRC, Alzheimer’s Research UK and the Alzheimer’s Society), Roger de Spoelberch Foundation, Alzheimer’s Research UK, The Tau Consortium, Cambridge Centre for Parkinson-Plus, National Institute for Health Research Cambridge Biomedical Research Centre (D.C.R.), Cambridge Commonwealth, European & International Trust (to AD, SK, and RP); Romanian grant of Ministry of Research and Innovation CNCS –UEFISCDI, project

References (240)

  • F.M. Menzies et al.

    Autophagy and neurodegeneration: pathogenic mechanisms and therapeutic opportunities

    Neuron

    (2017)
  • C.F. Bento et al.

    Mammalian autophagy: how does it work?

    Annu. Rev. Biochem.

    (2016)
  • B. Ravikumar et al.

    Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease

    Nat. Genet.

    (2004)
  • J.L. Webb et al.

    Alpha-Synuclein is degraded by both autophagy and the proteasome

    J. Biol. Chem.

    (2003)
  • V. Schaeffer et al.

    Stimulation of autophagy reduces neurodegeneration in a mouse model of human tauopathy

    Brain

    (2012)
  • A.R. Winslow et al.

    The Parkinson disease protein alpha-synuclein inhibits autophagy

    Autophagy

    (2011)
  • A. Ashkenazi et al.

    Polyglutamine tracts regulate beclin 1-dependent autophagy

    Nature

    (2017)
  • F.M. Menzies et al.

    Compromised autophagy and neurodegenerative diseases

    Nat. Rev. Neurosci.

    (2015)
  • D.C. Rubinsztein et al.

    Mechanisms of autophagosome biogenesis

    Curr. Biol.

    (2012)
  • X.H. Liang et al.

    Induction of autophagy and inhibition of tumorigenesis by beclin 1

    Nature

    (1999)
  • R.C. Russell et al.

    ULK1 induces autophagy by phosphorylating Beclin-1 and activating VPS34 lipid kinase

    Nat. Cell Biol.

    (2013)
  • N. Fujita et al.

    An Atg4B mutant hampers the lipidation of LC3 paralogues and causes defects in autophagosome closure

    Mol. Biol. Cell

    (2008)
  • S. Kimura et al.

    Dynein-dependent movement of autophagosomes mediates efficient encounters with lysosomes

    Cell Struct. Funct.

    (2008)
  • N. Hosokawa et al.

    Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy

    Mol. Biol. Cell

    (2009)
  • C. Settembre et al.

    TFEB links autophagy to lysosomal biogenesis

    Science

    (2011)
  • R.H. Brown et al.

    Amyotrophic lateral sclerosis

    N. Engl. J. Med.

    (2017)
  • M. Jimenez-Sanchez et al.

    Huntington's disease: mechanisms of pathogenesis and therapeutic strategies

    Cold Spring Harb Perspect Med

    (2017)
  • B. Dehay et al.

    Targeting alpha-synuclein for treatment of Parkinson's disease: mechanistic and therapeutic considerations

    Lancet Neurol.

    (2015)
  • B. De Strooper et al.

    The cellular Phase of alzheimer's disease

    Cell

    (2016)
  • V.M. Lee et al.

    Neurodegenerative tauopathies

    Annu. Rev. Neurosci.

    (2001)
  • M. Hansen et al.

    Autophagy as a promoter of longevity: insights from model organisms

    Nat. Rev. Mol. Cell Biol.

    (2018)
  • F. Pickford et al.

    The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice

    J. Clin. Investig.

    (2008)
  • R.A. Nixon

    The role of autophagy in neurodegenerative disease

    Nat. Med.

    (2013)
  • M. Shibata et al.

    Regulation of intracellular accumulation of mutant Huntingtin by Beclin 1

    J. Biol. Chem.

    (2006)
  • T.T. Rohn et al.

    Depletion of Beclin-1 due to proteolytic cleavage by caspases in the Alzheimer's disease brain

    Neurobiol. Dis.

    (2011)
  • M. Brehme et al.

    A chaperome subnetwork safeguards proteostasis in aging and neurodegenerative disease

    Cell Rep.

    (2014)
  • M. Pavel et al.

    CCT complex restricts neuropathogenic protein aggregation via autophagy

    Nat. Commun.

    (2016)
  • G. Jun et al.

    Meta-analysis confirms CR1, CLU, and PICALM as alzheimer disease risk loci and reveals interactions with APOE genotypes

    Arch. Neurol.

    (2010)
  • K. Ando et al.

    Clathrin adaptor CALM/PICALM is associated with neurofibrillary tangles and is cleaved in Alzheimer's brains

    Acta Neuropathol.

    (2013)
  • K. Moreau et al.

    PICALM modulates autophagy activity and tau accumulation

    Nat. Commun.

    (2014)
  • J.H. Lee et al.

    Lysosomal proteolysis and autophagy require presenilin 1 and are disrupted by Alzheimer-related PS1 mutations

    Cell

    (2010)
  • J.K. Lee et al.

    Acid sphingomyelinase modulates the autophagic process by controlling lysosomal biogenesis in Alzheimer's disease

    J. Exp. Med.

    (2014)
  • Z.S. Ji et al.

    Reactivity of apolipoprotein E4 and amyloid beta peptide: lysosomal stability and neurodegeneration

    J. Biol. Chem.

    (2006)
  • A.M. Cataldo et al.

    Endocytic pathway abnormalities precede amyloid beta deposition in sporadic Alzheimer's disease and Down syndrome: differential effects of APOE genotype and presenilin mutations

    Am. J. Pathol.

    (2000)
  • Y. Shi et al.

    ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy

    Nature

    (2017)
  • W. Xu et al.

    Amyloid precursor protein-mediated endocytic pathway disruption induces axonal dysfunction and neurodegeneration

    J. Clin. Investig.

    (2016)
  • B. Boland et al.

    Autophagy induction and autophagosome clearance in neurons: relationship to autophagic pathology in Alzheimer's disease

    J. Neurosci.

    (2008)
  • R.A. Nixon et al.

    Extensive involvement of autophagy in Alzheimer disease: an immuno-electron microscopy study

    J. Neuropathol. Exp. Neurol.

    (2005)
  • P. Ejlerskov et al.

    Genetic enhancement of macroautophagy in vertebrate models of neurodegenerative diseases

    Neurobiol. Dis.

    (2019)
  • A. Lopez et al.

    A152T tau allele causes neurodegeneration that can be ameliorated in a zebrafish model by autophagy induction

    Brain

    (2017)
  • T. Jiang et al.

    Temsirolimus attenuates tauopathy in vitro and in vivo by targeting tau hyperphosphorylation and autophagic clearance

    Neuropharmacology

    (2014)
  • S. Ozcelik et al.

    Rapamycin attenuates the progression of tau pathology in P301S tau transgenic mice

    PLoS One

    (2013)
  • P. Spilman et al.

    Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-beta levels in a mouse model of Alzheimer's disease

    PLoS One

    (2010)
  • Z. Berger et al.

    Rapamycin alleviates toxicity of different aggregate-prone proteins

    Hum. Mol. Genet.

    (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)
  • M. Renna et al.

    Chemical inducers of autophagy that enhance the clearance of mutant proteins in neurodegenerative diseases

    J. Biol. Chem.

    (2010)
  • B.J. DeBosch et al.

    Trehalose inhibits solute carrier 2A (SLC2A) proteins to induce autophagy and prevent hepatic steatosis

    Sci. Signal.

    (2016)
  • J.A. Rodriguez-Navarro et al.

    Trehalose ameliorates dopaminergic and tau pathology in parkin deleted/tau overexpressing mice through autophagy activation

    Neurobiol. Dis.

    (2010)
  • E. Luengo et al.

    Pharmacological doses of melatonin impede cognitive decline in tau-related Alzheimer models, once tauopathy is initiated, by restoring the autophagic flux

    J. Pineal Res.

    (2019)
  • Y. Hashimoto et al.

    A rescue factor abolishing neuronal cell death by a wide spectrum of familial Alzheimer's disease genes and Abeta

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

    (2001)
  • Cited by (148)

    View all citing articles on Scopus

    Joint first authors.

    View full text