Review
Nucleocytoplasmic transport defects in neurodegeneration — Cause or consequence?

https://doi.org/10.1016/j.semcdb.2019.05.020Get rights and content

Abstract

Defects in nucleocytoplasmic transport have been associated with several neurodegenerative disorders and, in particular, the formation of pathological protein aggregates characteristic for the respective disease. However, whether impaired nucleocytoplasmic transport is a consequence of such aggregates or rather contributes to their formation is still mostly unclear.

In this review, we summarize recent findings how both soluble and stationary components of the nucleocytoplasmic transport machinery are altered in neurodegenerative diseases, in particular amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), Alzheimer’s disease (AD) and Huntington’s disease (HD). We discuss the functional significance of the observed defects for nucleocytoplasmic transport of proteins and mRNAs. Moreover, we highlight interesting parallels observed in physiological ageing and the premature ageing syndrome progeria and propose that they that might provide mechanistic insights also for neurodegenerative processes.

Introduction

Neurodegeneration, the progressive dysfunction and eventual loss of specific nerve cells, is characterized by distinct cytosolic or nuclear protein deposits that are considered to be pathological hallmarks of the respective neurodegenerative disorder (Fig. 1). For example in most ALS (amyotrophic lateral sclerosis) and FTD (frontotemporal dementia) patients, the RNA binding proteins (RBPs) TDP43 or, more rarely, FUS are depleted from the nucleus and aggregate in the cytoplasm of neurons and glia cells in the affected brain regions [1]. ALS and FTD are part of a continuous disease spectrum and are linked by similar molecular dysfunctions [2,3]. For instance, they can be caused by the same genetic defects, e.g. a hexanucleotide (G4C2) repeat expansion in the first intron of the C9ORF72 gene (C9-ALS/FTD) [4,5]. While this repeat expansion also results in reduced expression of the C9ORF72 protein, disease-related pathology has largely been attributed to either RNA foci formed by sense and antisense repeat RNA (C9-RNA) or to inclusions formed by dipeptide repeat proteins (DPRs), namely polyGA, -GP, -GR, -PA and -PR, generated by ATG-independent translation from both sense and antisense C9-RNA [6].

A subset of FTD patients do not display RBP aggregates, but presents with intracellular aggregates of the microtubule-binding protein Tau in so-called neurofibrillary tangles [7]. Tau tangles, together with extracellular Aβ plaques, are also a neuropathological hallmark of Alzheimer’s disease (AD) [8]. Finally, CAG nucleotide repeat expansions result in expanded polyglutamine (polyQ) stretches in proteins such as Huntingtin (HTT), ataxins or the androgen receptor (AR) and subsequent intracellular aggregation of these proteins, and are associated with Huntington disease (HD), spinocerebellar ataxias (SCA) and spinobulbar muscular atrophy (SBMA), respectively [9]. How protein aggregates in various neurodegenerative disorders arise, and how they contribute to the pathogenic process of neurodegeneration, is currently not well understood.

In recent years, nucleocytoplasmic transport has gained increasing attention in neurodegeneration research, due to some prominent links between disease-associated protein aggregates and components of the nucleocytoplasmic transport machinery: First ALS-causing mutations in the nuclear localization signal (NLS) of FUS were identified, resulting in cytoplasmic mislocalization and accumulation of FUS in stress granules (SGs) [[10], [11], [12]]. This has led to the hypothesis that impaired nuclear import of disease-linked proteins may be a key ALS pathomechanism [13]. This hypothesis was further supported by several independent genetic screens for modulators of cellular toxicity, as well as proteomic studies that identified many components of the nucleocytoplasmic transport machinery to be associated with C9-ALS/FTD [[14], [15], [16], [17], [18]]. Recently, observations of nucleocytoplasmic transport defects in AD and HD [[19], [20], [21]] with some resemblance to physiological ageing [[22], [23], [24]] have further stimulated the discussion whether disturbances in nucleocytoplasmic transport may play a more global role in neurodegenerative processes than previously anticipated.

In this review, we summarize the status quo of research on nucleocytoplasmic transport in neurodegenerative diseases, in particular ALS/FTD, AD and HD. We will discuss whether nucleocytoplasmic transport defects are actually upstream of pathological protein deposits, contributing to their formation, or rather downstream, caused by the presence of pathological protein aggregates. Furthermore, we will highlight similarities between specific features of neurodegenerative disorders and physiological ageing, and discuss recent data obtained in progeria research that might provide interesting mechanistic insights into nucleocytoplasmic transport defects in neurodegenerative diseases. Overall, we hope to point out potential gaps in our current understanding of how nucleocytoplasmic transport defects are linked to neurodegeneration, which will require more detailed analysis in the future.

Section snippets

Evidence for impaired nucleocytoplasmic transport in neurodegenerative disorders

A number of nuclear proteins, such as RBPs or transcription factors, have been reported to mislocalize and aggregate in the cytoplasm of post mortem brain or spinal cord in diverse neurodegenerative disorders (Fig. 1; [4,5,7,9,11,12,14,20,[25], [26], [27], [28], [29], [30], [31], [32], [33], [34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45], [46], [47], [48]]). However, often it is unclear whether this is directly linked to impaired nucleocytoplasmic transport, as such

The role of the nuclear pore complex in neurodegeneration

As the NPC mediates controlled, bidirectional transport of proteins, RNA and complexes thereof between nucleus and cytoplasm, it is an obvious point of interest in search of mechanisms underlying nucleocytoplasmic transport defects in neurodegeneration.

NPCs are large multiprotein complexes (∼120 MDa in humans) composed of ∼30 individual proteins in multiple copies, so called nucleoporins (Nups) [[55], [56], [57], [58]]. The overall NPC architecture with its symmetrical core and asymmetrically

The role of NTRs in neurodegeneration

Nuclear transport receptors (NTRs), also called karyopherins, can be classified as importins or exportins, depending on the direction of cargo transport, even though some NTRs can function bidirectionally [56,128]. With ∼20 members, the Impβ superfamily is the largest class of NTRs and its members mediate import and export of both proteins and specific kinds of RNAs. NTRs of the Impβ-superfamily are ubiquitiously expressed, however their expression levels vary during development and between

Changes to the Ran system - misregulated regulators?

The differential concentration of RanGTP/GDP between nucleus and cytoplasm, generally referred to as RanGTP gradient, conveys directionality to nucleocytoplasmic transport and is maintained by spatial separation of its upstream regulators. In the nucleus, exchange of GDP to GTP is catalyzed by the chromatin-associated RanGEF, RCC1 [163]. On the cytoplasmic side, GTP hydrolysis is promoted by the RanGTPase activating protein, RanGAP, together with its accessory proteins RanBP1 or Nup358/RanBP2 [

Perspectives and open questions

Many lines of evidence indicate that nucleocytoplasmic transport is affected in several neurodegenerative diseases, however differences in the underlying mechanisms and molecular details seem to exist between distinct disorders. In order to dissect the individual pathogenic mechanisms and develop therapeutic tools, it will be essential to carefully examine the causal and temporal relationship of changes to the nucleocytoplasmic transport machinery in detail. In particular, it needs to be

Acknowledgements

We thank Lara Gruijs da Silva for helpful discussions and Dr. Ralph Kehlenbach for critical reading of the manuscript. This work was funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation; Emmy Noether grant DO 1804/1-1; DO1804/3-1) and under Germany’s Excellence Strategy within the framework of the Munich Cluster for Systems Neurology (EXC 2145 SyNergy – ID 390857198). We also acknowledge support from the Fritz Thyssen Foundation.

References (182)

  • N. Shibata

    Cu/Zn superoxide dismutase-like immunoreactivity in Lewy body-like inclusions of sporadic amyotrophic lateral sclerosis

    Neurosci. Lett.

    (1994)
  • A. Dickmanns et al.

    Nuclear pore complexes and nucleocytoplasmic transport: from structure to function to disease

    Int. Rev. Cell Mol. Biol.

    (2015)
  • K.E. Knockenhauer et al.

    The nuclear pore complex as a flexible and dynamic gate

    Cell

    (2016)
  • S. Frey et al.

    A saturated FG-repeat hydrogel can reproduce the permeability properties of nuclear pore complexes

    Cell

    (2007)
  • B.B. Hulsmann et al.

    The permeability of reconstituted nuclear pores provides direct evidence for the selective phase model

    Cell

    (2012)
  • M.P. Rout

    Virtual gating and nuclear transport: the hole picture

    Trends Cell Biol.

    (2003)
  • B.H. Toyama

    Identification of long-lived proteins reveals exceptional stability of essential cellular structures

    Cell

    (2013)
  • A.W. Folkmann

    Gle1 functions during mRNA export in an oligomeric complex that is altered in human disease

    Cell

    (2013)
  • J. Shang

    Aberrant distributions of nuclear pore complex proteins in ALS mice and ALS patients

    Neuroscience

    (2017)
  • T. Van Langenhove et al.

    The molecular basis of the frontotemporal lobar degeneration-amyotrophic lateral sclerosis spectrum

    Ann. Med.

    (2012)
  • R. Balendra et al.

    C9orf72-mediated ALS and FTD: multiple pathways to disease

    Nat. Rev. Neurol.

    (2018)
  • L.G. Bodea

    Tau physiology and pathomechanisms in frontotemporal lobar degeneration

    J. Neurochem.

    (2016)
  • J.C. Polanco

    Amyloid-beta and tau complexity – towards improved biomarkers and targeted therapies

    Nat. Rev. Neurol.

    (2018)
  • H.Y. Zoghbi et al.

    Glutamine repeats and neurodegeneration

    Annu. Rev. Neurosci.

    (2000)
  • D. Dormann

    ALS-associated fused in sarcoma (FUS) mutations disrupt Transportin-mediated nuclear import

    EMBO J.

    (2010)
  • T.J. Kwiatkowski

    Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis

    Science

    (2009)
  • C. Vance

    Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6

    Science

    (2009)
  • S. Boeynaems

    Drosophila screen connects nuclear transport genes to DPR pathology in c9ALS/FTD

    Sci. Rep.

    (2016)
  • B.D. Freibaum

    GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport

    Nature

    (2015)
  • A. Jovicic

    Modifiers of C9orf72 dipeptide repeat toxicity connect nucleocytoplasmic transport defects to FTD/ALS

    Nat. Neurosci.

    (2015)
  • K.H. Lee

    C9orf72 dipeptide repeats impair the assembly, dynamics, and function of membrane-less organelles

    Cell

    (2016)
  • K. Zhang

    The C9orf72 repeat expansion disrupts nucleocytoplasmic transport

    Nature

    (2015)
  • B. Eftekharzadeh

    Tau protein disrupts nucleocytoplasmic transport in Alzheimer’s disease

    Neuron

    (2018)
  • F. Gasset-Rosa

    Polyglutamine-expanded huntingtin exacerbates age-related disruption of nuclear integrity and nucleocytoplasmic transport

    Neuron

    (2017)
  • J.C. Grima

    Mutant huntingtin disrupts the nuclear pore complex

    Neuron

    (2017)
  • C. Amador-Ortiz

    TDP-43 immunoreactivity in hippocampal sclerosis and Alzheimer’s disease

    Ann. Neurol.

    (2007)
  • U. Bichelmeier

    Nuclear localization of ataxin-3 is required for the manifestation of symptoms in SCA3: in vivo evidence

    J. Neurosci.

    (2007)
  • L.I. Bruijn

    Aggregation and motor neuron toxicity of an ALS-linked SOD1 mutant independent from wild-type SOD1

    Science

    (1998)
  • M. DiFiglia

    Aggregation of huntingtin in neuronal intranuclear inclusions and dystrophic neurites in brain

    Science

    (1997)
  • K. Forsberg

    Novel antibodies reveal inclusions containing non-native SOD1 in sporadic ALS patients

    PLoS One

    (2010)
  • L.M. Gittings

    Heterogeneous nuclear ribonucleoproteins R and Q accumulate in pathological inclusions in FTLD-FUS

    Acta Neuropathol. Commun.

    (2019)
  • H. Hartmann

    Proteomics and C9orf72 neuropathology identify ribosomes as poly-GR/PR interactors driving toxicity

    Life Sci. Alliance

    (2018)
  • Y.D. Ke

    Tau-mediated nuclear depletion and cytoplasmic accumulation of SFPQ in Alzheimer’s and Pick’s disease

    PLoS One

    (2012)
  • M. Li

    Nuclear inclusions of the androgen receptor protein in spinal and bulbar muscular atrophy

    Ann. Neurol.

    (1998)
  • T. Lu

    REST and stress resistance in ageing and Alzheimer’s disease

    Nature

    (2014)
  • K. Mori

    hnRNP A3 binds to GGGGCC repeats and is a constituent of p62-positive/TDP43-negative inclusions in the hippocampus of patients with C9orf72 mutations

    Acta Neuropathol.

    (2013)
  • M. Neumann

    A new subtype of frontotemporal lobar degeneration with FUS pathology

    Brain

    (2009)
  • M. Neumann

    Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis

    Science

    (2006)
  • M. Neumann

    Transportin 1 accumulates specifically with FET proteins but no other transportin cargos in FTLD-FUS and is absent in FUS inclusions in ALS with FUS mutations

    Acta Neuropathol.

    (2012)
  • F.C. Nucifora

    Interference by huntingtin and atrophin-1 with cbp-mediated transcription leading to cellular toxicity

    Science

    (2001)
  • Cited by (53)

    • ALS-associated KIF5A mutations abolish autoinhibition resulting in a toxic gain of function

      2022, Cell Reports
      Citation Excerpt :

      We found that KIF5AR1007K iMNs had more cytoplasmic RAN than the isogenic control (Figures 6H and 6I), demonstrating that mutant KIF5A perturbs NCT. This is not surprising as dysregulation of NCT has been implicated in several neurodegenerative diseases, including ALS, FTD, AD, and HD (Ding and Sepehrimanesh, 2021; Hutten and Dormann, 2020). Neurodegenerative diseases represent a complex set of disorders in which there is dire need for improved therapeutics.

    View all citing articles on Scopus
    View full text