Elsevier

Ageing Research Reviews

Volume 64, December 2020, 101172
Ageing Research Reviews

Review
Insights into disease mechanisms and potential therapeutics for C9orf72-related amyotrophic lateral sclerosis/frontotemporal dementia

https://doi.org/10.1016/j.arr.2020.101172Get rights and content

Highlights

  • Loss- and gain-of-function mechanisms are involved in C9-FTD/ALS.

  • Different cellular downstream pathways are impaired in C9-FTD/ALS.

  • Several disease models have provided new insights into the pathogenic mechanisms.

  • Biochemical and neuroimaging biomarkers may represent useful tools for C9-ALS/FTD patients.

  • Therapeutic strategies targeting C9orf72 repeats are promising for clinical use.

Abstract

In 2011, a hexanucleotide repeat expansion (HRE) in the noncoding region of C9orf72 was associated with the most frequent genetic cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). The main pathogenic mechanisms in C9-ALS/FTD are haploinsufficiency of the C9orf72 protein and gain of function toxicity from bidirectionally-transcribed repeat-containing RNAs and dipeptide repeat proteins (DPRs) resulting from non-canonical RNA translation. Additionally, abnormalities in different downstream cellular mechanisms, such as nucleocytoplasmic transport and autophagy, play a role in pathogenesis. Substantial research efforts using in vitro and in vivo models have provided valuable insights into the contribution of each mechanism in disease pathogenesis. However, conflicting evidence exists, and a unifying theory still lacks.

Here, we provide an overview of the recently published literature on clinical, neuropathological and molecular features of C9-ALS/FTD. We highlight the supposed neuronal role of C9orf72 and the HRE pathogenic cascade, mainly focusing on the contribution of RNA foci and DPRs to neurodegeneration and discussing the several downstream mechanisms. We summarize the emerging biochemical and neuroimaging biomarkers, as well as the potential therapeutic approaches. Despite promising results, a specific disease-modifying treatment is still not available to date and greater insights into disease mechanisms may help in this direction.

Introduction

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder affecting upper and lower motor neurons (MNs) that causes progressive and relentless muscular weakness and atrophy leading to death for respiratory insufficiency. Frontotemporal dementia (FTD) is the most common form of dementia before 65 years of age and is characterized by relevant changes in social behavior and personality or language disturbance, due to degeneration of the frontal and/or temporal lobes. The term frontotemporal lobar degeneration (FTLD) denotes a neuropathological spectrum encompassing different neurodegenerative disorders, such as ALS, FTD and various combinations of them. Although the majority of these clinical syndromes are sporadic, a conspicuous part of them (ranging from 10 to 25 % in FTD and 5–10 % in ALS) is familial.

In 2011 De Jesus-Hernandez et al. and Renton et al. independently discovered that the hexanucleotide GGGGCC (G4C2) expansion in a non-coding region of Chromosome 9 Open reading frame 72 (C9orf72) gene on chromosome 9 is the most frequent genetic cause of both sporadic and familial forms of ALS and FTD in Western countries (DeJesus-Hernandez et al., 2011; Renton et al., 2011). The clinical phenotypes associated with C9orf72 hexanucleotide repeat expansion (HRE) are markedly heterogeneous (Woollacott and Mead, 2014), the number of repeats and the clinical presentation are not correlated and HRE length is highly variable in different tissues of the same patient and in parental offspring transmission.

Although the function of C9orf72 protein is not completely understood, both primary neurons from patients with C9orf72-related ALS/FTD (C9-ALS/FTD) and studies in in vitro and in vivo C9-models have improved our understanding of the etiology and pathogenesis of C9-ALS/FTD. Three possible, but not mutually exclusive pathomechanisms have emerged and are still debated in the field: loss-of-function due to C9orf72 protein haploinsufficiency, gain of toxic function from repeat-containing RNAs, also known as RNA foci, generated by bidirectional transcription of HRE, and toxic gain-of-function of dipeptide repeat proteins (DPRs) translated from sense and antisense HRE transcripts by non-canonical translation of repetitive RNAs (Balendra and Isaacs, 2018). Further, alterations in various downstream cellular pathways, such as nucleocytoplasmic transport (NCT), RNA metabolism, ubiquitin-proteasome system (UPS) and autophagy, vesicle trafficking, DNA damage, and mitochondrial metabolism, might be involved in disease pathogenesis.

The precise contribution of each mechanism into C9-ALS/FTD pathogenesis is still controversial, but they all likely play a role to some extent. The discovery of C9orf72 as the most common genetic cause of ALS/FTD has prompted the research towards an HRE-targeted therapy, opening the path for new promising pharmacologic approaches. However, though a phase I clinical trial on antisense oligonucleotides (ASOs) directed on HRE is now ongoing, an efficacious treatment for C9-ALS/FTD still lacks.

Here, we provide a literature overview on clinical, pathological and molecular aspects of C9-ALS/FTD. We report recent epidemiological data, clinical features and neuropathology underlying C9-ALS/FTD, highlighting the main similarities and differences between C9-ALS/FTD and sporadic form of ALS (sALS). We describe the gene structure and the supposed role of C9orf72 protein in cellular machinery. Through the description of neuropathological findings and in vitro and in vivo studies, we focus on the main three mechanisms involved in disease pathogenesis, investigating proofs and controversies regarding the contribution of each of them in driving neurodegeneration, and widely exploring the downstream cellular pathways. Then, we highlight the most recently investigated biochemical, structural and functional biomarkers to assess disease progression and drug efficacy in clinical trials. Finally, we provide a synopsis of the possible therapeutic approaches in C9-ALS/FTD, including ASOs specifically targeting the C9orf72 HRE, RNA interference, small molecules, genome editing techniques and DPRs-directed strategies.

We are confident that a better comprehension of the molecular mechanisms underlying C9-ALS/FTD pathogenesis would provide major and promising advances in specific disease-modifying treatments.

Section snippets

Epidemiology and clinical features

HRE in C9orf72 is the most common genetic cause of ALS and FTD all over the world and in particular in the Caucasian population, while it is very infrequent in the Asiatic population. Large population studies conducted by Majounie and colleagues in 2012 have shown that the prevalence of G4C2 repeat expansion in C9orf72 accounts for 5–7% of sporadic ALS and FTD forms in white Europeans, Americans, and Australians (Majounie et al., 2012b). The pathogenic expansion is more frequent in white (7%)

C9orf72 gene and the G4C2 repeat expansion

The C9orf72 gene is placed on the short arm of chromosome 9 (9p21), contains twelve exons and generates three transcription variants by using alternative transcription start and termination sites (Fig. 1). The HRE is located in a non-coding portion of the gene, in the first intron of variants 1 and 3, and within the promoter region of the variant 2. Despite the intronic location of the G4C2 repeats in variants 1 and 3, they are likely retained in the mature messenger RNA (mRNA) and successfully

Protein function

C9orf72-related transcript variants encode for two different protein isoforms. Transcript variants 2 and 3 are translated into the full-length C9orf72 protein (481 amino acids), and variant 1 yields a predicted short protein (222 amino acids) (Fig. 1) (Woollacott and Mead, 2014). It is not clear whether the predicted short isoform C9orf72 is present in nature. Xiao et al. have shown a decreased short isoform expression in nuclear membranes of C9-ALS/FTD cases compared with controls, by using an

Neuropathology

Most ALS and approximately half of FTD cases show TAR-DNA binding protein 43 (TDP-43) inclusions in the nucleus and cytoplasm of neurons and oligodendroglia (Neumann et al., 2006), and are usually acknowledged as FTLD-TDP. These aggregates display abnormal TDP-43 phosphorylation and ubiquitination and truncated C-terminal fragments of TDP-43 (Arai et al., 2006; Neumann et al., 2006). Neuropathology and genetic alterations are correlated and C9-ALS/FTD cases are associated with type A and type B

Mechanisms of neurodegeneration

Three different but not mutually exclusive mechanisms contribute to C9orf72-mediated neurodegeneration: loss of function of C9orf72 protein due to haploinsufficiency, toxic gain of function from sense and antisense RNA foci, which are bidirectionally transcribed from HRE into repetitive RNAs, and toxic gain of function from DPRs, derived from repetitive RNA non-canonical translation. All these processes play a role in C9-ALS/FTD pathogenesis to some extent, and they are likely responsible for

Downstream mechanisms

In addition to C9orf72 loss- and gain-of-function mechanism, various studies have focused on downstream cellular alterations resulting from the HRE pathogenic cascade, including altered nucleocytoplasmic transport and RNA metabolism, disrupted proteostasis as well as other cellular functions. Researchers have used multiple human and non-human model systems for this purpose (Fig. 3).

Biochemical biomarkers

Biomarkers in ALS may help to make an earlier diagnosis, to enroll patients in clinical trials and to monitor the efficacy of therapeutic interventions. Biochemical biomarkers include DPRs, neurofilaments (Nfs) and other proteins that are related to the pathogenesis of C9-ALS/FTD and can be measured in biofluids or biological tissues.

CSF poly-GP levels discriminate symptomatic and pre-symptomatic carriers from non-carriers, thus they may be useful as pharmacodynamic biomarkers (Floeter and

Therapeutics

To date, no cure exists for C9-ALS/FTD. Different studies using ASO technology, small molecules, CRISPR-Cas9 as well as RNA interference (RNAi) strategies have targeted various pathogenic pathways, including C9orf72 RNA and DNA, DPRs, TDP-43 and downstream mechanisms (Table 2).

Closing remarks

Since the identification of C9orf72 repeat expansion, many studies have focused on unraveling the underlying pathogenic mechanisms in C9-ALS/FTD. At the same time, several disease models in vitro and in vivo have provided insights into the loss/gain of function-related neurotoxicity, as well as the downstream cellular function abnormalities. Despite thorough investigations of both biomarkers and new therapies have proceeded enthusiastically, disease-modifying therapies for C9-ALS/FTD still

Author statement

All authors have approved the final draft of the article.

Funding

This study was funded by Italian Ministry Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico Ricerca Corrente 2020 to NB and GPC.

CRediT authorship contribution statement

Delia Gagliardi: Conceptualization, Investigation, Writing - original draft. Gianluca Costamagna: Conceptualization, Investigation, Writing - original draft. Michela Taiana: Investigation, Writing - review & editing. Luca Andreoli: Visualization. Fabio Biella: Investigation, Visualization. Margherita Bersani: Visualization. Nereo Bresolin: Supervision. Giacomo Pietro Comi: Supervision. Stefania Corti: Conceptualization, Writing - review & editing, Supervision.

Declaration of Competing Interest

The authors reported no declarations of interest.

Acknowledgments

We gratefully thank the Associazione Centro Dino Ferrari for its support. Regione Lombardia TRANS-ALS and RF-2013-02355764/Italian Ministry of Health to GPC is gratefully acknowledged. Italian Ministry of health "RF-2018-12366357 grant to SC is gratefully acknowledged.

References (275)

  • A.E. Conicella et al.

    ALS mutations disrupt phase separation mediated by α-Helical structure in the TDP-43 low-complexity C-Terminal domain

    Structure

    (2016)
  • A.N. Coyne et al.

    Post-transcriptional inhibition of Hsc70-4/HSPA8 expression leads to synaptic vesicle cycling defects in multiple models of ALS

    Cell Rep.

    (2017)
  • R. Cristofani et al.

    The small heat shock protein B8 (HSPB8) efficiently removes aggregating species of dipeptides produced in C9ORF72-related neurodegenerative diseases

    Cell Stress Chaperones

    (2018)
  • M. DeJesus-Hernandez et al.

    Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS

    Neuron

    (2011)
  • C.J. Donnelly et al.

    RNA toxicity from the ALS/FTD C9ORF72 expansion is mitigated by antisense intervention

    Neuron

    (2013)
  • M.M. Fay et al.

    ALS/FTD-associated C9ORF72 repeat RNA promotes phase transitions in vitro and in cells

    Cell Rep.

    (2017)
  • M.K. Floeter et al.

    Longitudinal imaging in C9orf72 mutation carriers: relationship to phenotype

    Neuroimage Clin.

    (2016)
  • I. Gijselinck et al.

    A C9orf72 promoter repeat expansion in a Flanders-Belgian cohort with disorders of the frontotemporal lobar degeneration-amyotrophic lateral sclerosis spectrum: a gene identification study

    Lancet Neurol.

    (2012)
  • C. Akimoto et al.

    No GGGGCC-hexanucleotide repeat expansion in C9ORF72 in parkinsonism patients in Sweden

    Amyotroph. Lateral Scler. Frontotemporal Degener.

    (2013)
  • S. Almeida et al.

    Modeling key pathological features of frontotemporal dementia with C9ORF72 repeat expansion in iPSC-derived human neurons

    Acta Neuropathol.

    (2013)
  • S. Al-Sarraj et al.

    p62 positive, TDP-43 negative, neuronal cytoplasmic and intranuclear inclusions in the cerebellum and hippocampus define the pathology of C9orf72-linked FTLD and MND/ALS

    Acta Neuropathol.

    (2011)
  • Joseph Amick et al.

    C9orf72: At the intersection of lysosome cell biology and neurodegenerative disease

    Traffic

    (2017)
  • J. Amick et al.

    C9orf72 binds SMCR8, localizes to lysosomes, and regulates mTORC1 signaling

    MBoC

    (2016)
  • Y. Aoki et al.

    C9orf72 and RAB7L1 regulate vesicle trafficking in amyotrophic lateral sclerosis and frontotemporal dementia

    Brain

    (2017)
  • H.C. Archbold et al.

    TDP43 nuclear export and neurodegeneration in models of amyotrophic lateral sclerosis and frontotemporal dementia

    Sci. Rep.

    (2018)
  • S. Asamitsu et al.

    Perspectives for applying G-Quadruplex structures in neurobiology and neuropharmacology

    Int. J. Mol. Sci.

    (2019)
  • A. Atanasio et al.

    C9orf72 ablation causes immune dysregulation characterized by leukocyte expansion, autoantibody production, and glomerulonephropathy in mice

    Sci. Rep.

    (2016)
  • R.A.K. Atkinson et al.

    C9ORF72 expression and cellular localization over mouse development

    Acta Neuropathol. Commun.

    (2015)
  • A. Baborie et al.

    Accumulation of dipeptide repeat proteins predates that of TDP-43 in frontotemporal lobar degeneration associated with hexanucleotide repeat expansions in C9ORF72 gene

    Neuropathol. Appl. Neurobiol.

    (2015)
  • R. Balendra et al.

    C9orf72-mediated ALS and FTD: multiple pathways to disease

    Nat. Rev. Neurol.

    (2018)
  • P. Barschke et al.

    Different CSF protein profiles in amyotrophic lateral sclerosis and frontotemporal dementia with C9orf72 hexanucleotide repeat expansion

    J Neurol Neurosurg Psychiatry

    (2020)
  • R. Batra et al.

    Elimination of toxic microsatellite repeat expansion RNA by RNA-Targeting Cas9

    Cell

    (2017)
  • L.A. Becker et al.

    Therapeutic reduction of ataxin-2 extends lifespan and reduces pathology in TDP-43 mice

    Nature

    (2017)
  • V.V. Belzil et al.

    Reduced C9orf72 gene expression in c9FTD/ALS is caused by histone trimethylation, an epigenetic event detectable in blood

    Acta Neuropathol.

    (2013)
  • M. Benatar et al.

    Neurofilament light: a candidate biomarker of presymptomatic amyotrophic lateral sclerosis and phenoconversion

    Ann. Neurol.

    (2018)
  • A. Bertrand et al.

    Early cognitive, structural, and microstructural changes in presymptomatic C9orf72 carriers younger than 40 years

    JAMA Neurol.

    (2018)
  • V. Bhatia et al.

    BRCA2 prevents R-loop accumulation and associates with TREX-2 mRNA export factor PCID2

    Nature

    (2014)
  • G. Biffi et al.

    Visualization and selective chemical targeting of RNA G-quadruplex structures in the cytoplasm of human cells

    Nat. Chem.

    (2014)
  • B.F. Boeve et al.

    Characterization of frontotemporal dementia and/or amyotrophic lateral sclerosis associated with the GGGGCC repeat expansion in C9ORF72

    Brain

    (2012)
  • S. Boeynaems et al.

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

    Sci. Rep.

    (2016)
  • S. Boeynaems et al.

    Phase separation of C9orf72 dipeptide repeats perturbs stress granule dynamics

    Mol. Cell

    (2017)
  • M. Boivin et al.

    Reduced autophagy upon C9ORF72 loss synergizes with dipeptide repeat protein toxicity in G4C2 repeat expansion disorders

    EMBO J.

    (2020)
  • A. Boutajangout et al.

    Passive immunization targeting pathological phospho-tau protein in a mouse model reduces functional decline and clears tau aggregates from the brain

    J. Neurochem.

    (2011)
  • A. Burberry et al.

    Loss-of-function mutations in the C9ORF72 mouse ortholog cause fatal autoimmune disease

    Sci. Transl. Med.

    (2016)
  • E. Caverzasi et al.

    Gyrification abnormalities in presymptomatic c9orf72 expansion carriers

    J Neurol Neurosurg Psychiatry

    (2019)
  • W. Cheng et al.

    CRISPR-Cas9 screens identify the RNA helicase DDX3X as a repressor of C9ORF72 (GGGGCC)n repeat-associated Non-AUG translation

    Neuron

    (2019)
  • J. Chew et al.

    C9ORF72 repeat expansions in mice cause TDP-43 pathology, neuronal loss, and behavioral deficits

    Science

    (2015)
  • A. Chiò et al.

    Clinical characteristics of patients with familial amyotrophic lateral sclerosis carrying the pathogenic GGGGCC hexanucleotide repeat expansion of C9ORF72

    Brain

    (2012)
  • M. Chitiprolu et al.

    A complex of C9ORF72 and p62 uses arginine methylation to eliminate stress granules by autophagy

    Nat. Commun.

    (2018)
  • S.Y. Choi et al.

    C9ORF72-ALS/FTD-associated poly(GR) binds Atp5a1 and compromises mitochondrial function in vivo

    Nat. Neurosci.

    (2019)
  • Cited by (5)

    • Evolving markers in amyotrophic lateral sclerosis

      2023, Advances in Clinical Chemistry
    1

    These authors contributed equally to this work.

    View full text