Skip to main content

ORIGINAL RESEARCH article

Front. Cell. Neurosci., 15 December 2022
Sec. Cellular Neuropathology
Volume 16 - 2022 | https://doi.org/10.3389/fncel.2022.1050596

Telomere length analysis in amyotrophic lateral sclerosis using large-scale whole genome sequence data

Ahmad Al Khleifat1*† Alfredo Iacoangeli1,2† Ashley R. Jones1 Joke J. F. A. van Vugt3 Matthieu Moisse4,5 Aleksey Shatunov6,7 Ramona A. J. Zwamborn3 Rick A. A. van der Spek3 Johnathan Cooper-Knock8 Simon Topp1 Wouter van Rheenen3 Brendan Kenna3 Kristel R. Van Eijk3 Kevin Kenna3 Ross Byrne9 Victoria López10 Sarah Opie-Martin1 Atay Vural11 Yolanda Campos10 Markus Weber11,12 Bradley Smith1 Isabella Fogh1 Vincenzo Silani13,14 Karen E. Morrison15 Richard Dobson2,16 Michael A. van Es3 Russell L. McLaughlin9 Patrick Vourc’h17 Adriano Chio18,19 Philippe Corcia17,20 Mamede de Carvalho21 Marc Gotkine22 Monica Povedano Panades23 Jesus S. Mora24 Pamela J. Shaw8 John E. Landers25 Jonathan D. Glass26 Christopher E. Shaw1,27 Nazli Basak11 Orla Hardiman28,29 Wim Robberecht4,30 Philip Van Damme4,30 Leonard H. van den Berg3 Jan H. Veldink3 Ammar Al-Chalabi1,27* on behalf of the Project MinE Consortium
  • 1Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
  • 2Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
  • 3Department of Neurology, University Medical Center (UMC) Utrecht Brain Center, Utrecht University, Utrecht, Netherlands
  • 4Department of Neurosciences, Experimental Neurology, KU Leuven—University of Leuven, Leuven, Belgium
  • 5VIB Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
  • 6Institute of Medicine, North-Eastern Federal University, Yakutsk, Russia
  • 7Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
  • 8Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom
  • 9Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
  • 10Computational Biology Unit, Instituto de Salud Carlos III, Madrid, Spain
  • 11School of Medicine, Translational Medicine Research Center-NDAL, Koc University, Istanbul, Turkey
  • 12Neuromuscular Diseases Unit/ALS Clinic, Kantonsspital St. Gallen, St. Gallen, Switzerland
  • 13Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
  • 14Department of Pathophysiology and Transplantation, “Dino Ferrari” Center, Università degli Studi di Milano, Milan, Italy
  • 15Faculty of Medicine, Health and Life Sciences, Queen’s University Belfast, Belfast, United Kingdom
  • 16Institute of Health Informatics, University College London, London, United Kingdom
  • 17Centre SLA, CHRU de Tours, Tours, France
  • 18Department of Neuroscience, ALS Centre, University of Torino, Turin, Italy
  • 19Azienda Ospedaliera Citta della Salute e della Scienza, Turin, Italy
  • 20Federation des Centres SLA Tours and Limoges, LITORALS, Tours, France
  • 21Physiology Institute, Faculty of Medicine, Instituto de Medicina Molecular, University of Lisbon, Lisbon, Portugal
  • 22Department of Neurology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
  • 23Department of Neurology, Hospital Universitari de Bellvitge, Barcelona, Spain
  • 24Hospital San Rafael, Madrid, Spain
  • 25Department of Neurology, University of Massachusetts Medical School, Worcester, MA, United States
  • 26Department of Neurology, Center for Neurodegenerative Diseases, Emory University, Atlanta, GA, United States
  • 27King’s College Hospital, London, United Kingdom
  • 28Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
  • 29Department of Neurology, Beaumont Hospital, Dublin, Ireland
  • 30Department of Neurology, University Hospitals Leuven, Leuven, Belgium

Background: Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the loss of upper and lower motor neurons, leading to progressive weakness of voluntary muscles, with death following from neuromuscular respiratory failure, typically within 3 to 5 years. There is a strong genetic contribution to ALS risk. In 10% or more, a family history of ALS or frontotemporal dementia is obtained, and the Mendelian genes responsible for ALS in such families have now been identified in about 50% of cases. Only about 14% of apparently sporadic ALS is explained by known genetic variation, suggesting that other forms of genetic variation are important. Telomeres maintain DNA integrity during cellular replication, differ between sexes, and shorten naturally with age. Sex and age are risk factors for ALS and we therefore investigated telomere length in ALS.

Methods: Samples were from Project MinE, an international ALS whole genome sequencing consortium that includes phenotype data. For validation we used donated brain samples from motor cortex from people with ALS and controls. Ancestry and relatedness were evaluated by principal components analysis and relationship matrices of DNA microarray data. Whole genome sequence data were from Illumina HiSeq platforms and aligned using the Isaac pipeline. TelSeq was used to quantify telomere length using whole genome sequence data. We tested the association of telomere length with ALS and ALS survival using Cox regression.

Results: There were 6,580 whole genome sequences, reducing to 6,195 samples (4,315 from people with ALS and 1,880 controls) after quality control, and 159 brain samples (106 ALS, 53 controls). Accounting for age and sex, there was a 20% (95% CI 14%, 25%) increase of telomere length in people with ALS compared to controls (p = 1.1 × 10−12), validated in the brain samples (p = 0.03). Those with shorter telomeres had a 10% increase in median survival (p = 5.0×10−7). Although there was no difference in telomere length between sporadic ALS and familial ALS (p=0.64), telomere length in 334 people with ALS due to expanded C9orf72 repeats was shorter than in those without expanded C9orf72 repeats (p = 5.0×10−4).

Discussion: Although telomeres shorten with age, longer telomeres are a risk factor for ALS and worsen prognosis. Longer telomeres are associated with ALS.

Introduction

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease affecting motor neurons in the brain and spinal cord resulting in progressive paralysis and death, within three to five years, typically due to respiratory failure (Brown and Al-Chalabi, 2017; Hardiman et al., 2017). The first symptoms of weakness can occur in the bulbar innervated muscles, manifesting as difficulty with speech or swallowing, or in the spinal innervated muscles, manifesting as limb weakness or breathing difficulty. About 5% may have a frank frontotemporal dementia, and frontotemporal impairment is seen in up to 80% of people by the time King’s Stage 4 disease is reached (Roche et al., 2012; Crockford et al., 2018).

The last decade has seen substantial advances in our understanding of the genomic basis of ALS (van Rheenen et al., 2021; Hop et al., 2022) but a significant proportion of the genetic contribution to risk remains unexplained. This hidden heritability may be harbored in other types of genomic variation as well as in rare variants that may be unique to an affected individual or family (Al-Chalabi et al., 2010; McLaughlin et al., 2015).

Telomeres are repeated TTAGGG nucleotide sequences located at the ends of chromosomes and exist to maintain chromosomal structural integrity during cellular replication. They shorten naturally with age and differ in average length between the sexes (Muzumdar and Atzmon, 2012; Kong et al., 2013; Gardner et al., 2014); age and sex are also risk factors for ALS (McCombe and Henderson, 2010; Al-Chalabi and Hardiman, 2013; Westeneng et al., 2018). Telomere length is a marker for aging, chromosomal instability and DNA damage, and might therefore be relevant as a risk factor for ALS (Murnane, 2006; Conomos et al., 2012; Xu et al., 2013; Marzec et al., 2015).

Previously, in a pilot study in a UK cohort, we showed that longer telomeres might be associated with ALS when compared to age and sex-matched controls (Al Khleifat et al., 2019). We therefore sought to explore this finding in detail, using whole-genome sequence data from the Project MinE consortium (van Rheenen et al., 2018), a large international ALS genomics collaboration.

Materials and methods

Data sources and data extraction

Blood samples

Samples were from the international Project MinE whole genome sequencing consortium and derived from seven countries: the USA, Ireland, Belgium, the Netherlands, Spain, Turkey, and the United Kingdom (van Rheenen et al., 2018).

DNA was isolated from venous blood using standard methods. The DNA concentrations were set at 100 ng/ul as measured by a fluorimeter with the PicoGreen® dsDNA quantitation assay. DNA integrity was assessed using gel electrophoresis.

Post-mortem samples

Post-mortem motor cortex was from the MRC London Neurodegenerative Diseases Brain Bank based at the Institute of Psychiatry, Psychology and Neuroscience, King’s College London. Tissue was flash frozen stored at −80°C. 100 mg tissue blocks were excised. DNA was isolated from the same tissue block and sequenced. The study cohort consisted of 64 people with apparently sporadic ALS and 53 controls with no known neurological disease (controls below Hyperphosphorylated tau (HP-τ) in human brain tissue and BNE\Braak stage 2).

The 100 mg tissue blocks were divided to allow DNA purification. For each sample, a 25 mg tissue block for DNA was homogenized using a Qiagen PowerLyzer 24 Homogenizer. DNA was purified from the homogenate using the standard protocol from Qiagen’s DNeasy Blood and Tissue Mini Kit. DNA was quantified using PicoGreen (Quant-iT™ PicoGreen® dsDNA Reagent, ThermoFisher Scientific) and measured using a Spectromax Gemini XPS (Molecular Devices).

Library preparation and DNA sequencing

Library preparation was performed using the Illumina DNA Sample Preparation HT Kit alongside the Illumina SeqLab DNA PCR-Free Library Prep Guide. Libraries were then quantified using qPCR and evaluated using gel electrophoresis. All samples were sequenced using Illumina’s FastTrack services (San Diego, CA, USA). Some blood-derived DNA samples were sequenced using the Illumina HiSeq 2000 platform. Sequencing was 100 bp paired-end performed using PCR-free library preparations and targeted ∼40x coverage across each sample. Remaining blood-derived and all brain-derived DNA libraries were clustered onto flow cells using the Illumina cBot System, as per cBot System Guide using Illumina HiSeq X HD Paired End Cluster Kit reagents and sequenced on an Illumina HiSeqX with 151 bp paired-end runs using independent flow cell lanes and with a target minimum of 30x average coverage per sample. Binary sequence alignment/map formats (BAM) were generated for each individual. All the genomes were aligned with Isaac (Illumina) to hg19. The details of the Isaac alignment and variant calling pipelines are discussed in Project MinE design (van Rheenen et al., 2018) and the Isaac protocol (Raczy et al., 2013).

Determination of telomere length

TelSeq (Ding et al., 2014) was used to quantify telomere length using whole genome sequence data. Telomere lengths were estimated from reads, defined as repeats of more than seven TTAGGG motifs.

Statistical analysis

The effect of telomere length on ALS risk was tested using a multivariable linear regression model. To account for different sequencing platforms and population stratification, principal components of ancestry, center and technology platform were included as covariates. To assess the model, Pearson’s chi-squared test was used. Because telomere length correlates with age, we performed an additional test to examine the possibility that survival bias could affect the results. To do this, we also performed the analysis restricted to the subgroup of people with ALS onset below the median cohort age (62 years). As brain is composed of neurons which do not divide, as well as glia which do, we expected that the average telomere length in brain would be longer than in blood. Furthermore, nervous tissue is the target of the disease process rather than blood. We therefore additionally tested the effect of age on telomere length in brain tissue.

To determine if telomeres are lengthened in ALS, or simply shorten less rapidly than in controls, we analysed the effect of age on telomere length in each group using multivariable linear regression.

To assess the effect of covariates on telomere length affecting survival, we used Cox regression, controlling for age, sex and site of disease onset (bulbar or spinal), population stratification, principal components, center and sequencing platforms.

Repeat primed PCR and Expansion Hunter-v2.5.1 (Dolzhenko et al., 2017) were used to assay the hexanucleotide repeat expansion in the C9orf72 gene since this is a known risk factor for ALS and associates with survival.

Statistical tests were performed using IBM SPSS Statistics 24.0 (SPSS Inc., Armonk, NY, USA), and RStudio, R Foundation for Statistical Computing 3.4.1 (RStudio, Vienna, Austria).

Quality control

Quality control was performed separately on the genotyped data of each population as reported previously (van Rheenen et al., 2016; Supplementary Appendix 1).

Ethical approval

Informed consent was obtained from all participants in this project according to the ethical approval at each participating Project MinE site as previously described (van Rheenen et al., 2018).

Results

There were 6,580 whole genome sequences (4,515 from people with ALS and 2,065 from controls), reducing to 6,195 [4,315 (95.6%) from people with ALS and 1,880 (91%) from controls] after quality control, with minimum ∼25x coverage across each sample. The set was enriched for apparently sporadic ALS [4,236 compared with 79 with familial ALS (FALS)]. The male-female ratio was 2:1. Overall, 22 had ALS-frontotemporal dementia (ALS-FTD). Phenotypically, 37 had pure progressive bulbar palsy (PBP) and 68 and progressive muscular atrophy. There were 1,908 sequenced using the HiSeq2000 platform and 4,287 sequenced using the HiSeqX Illumina platform (Table 1). There were 344 people carrying an expanded C9orf72 hexanucleotide repeat, 334 with ALS and 10 without symptoms.

TABLE 1
www.frontiersin.org

Table 1. Detailed demographic features of the study population.

The mean telomere length in people with ALS was 5.5 kb, and in controls, 5.38 kb (Figure 1). Multivariable linear regression accounting for sex and age as covariates showed a mean 20% (95% CI 14, 25%) longer telomere length in people with ALS compared to controls (p = 1.1 × 10–12). Covariate analysis showed that regardless of disease status, females (p = 2.42 × 10–5) and younger people (p = 1.2 × 10–16) had on average longer telomeres (Table 2), confirming the results of earlier studies that telomere length reduces with age and females have on average longer telomeres.

FIGURE 1
www.frontiersin.org

Figure 1. Mean telomere length by age (A), sex (B), and disease status (C). Purple bars indicate 95% confidence intervals.

TABLE 2
www.frontiersin.org

Table 2. Telomere length comparison between people with amyotrophic lateral sclerosis (ALS) and healthy controls using a generalized linear model.

To assess if the observed longer telomere length in apparently sporadic ALS is also seen in familial ALS, we assessed telomere length in 79 people, not included in the main analysis, with a family history of ALS in a first degree relative (FALS). Multivariable linear regression after correcting for age and sex again showed a longer average telomere length in people with FALS than in controls (p = 2.0 × 10–16).

Examining the effect of age on telomere length in ALS and controls separately, showed that the rate of shortening by age is slower in ALS than in controls, suggesting it is not an active lengthening of telomeres in ALS (0.022% per year, p < 0.0001 vs. 0.012% per year, p < 0.0001) and also arguing against the possibility that telomeres are longer to start with in people who will later develop ALS. The rate of shortening was different in males and females, with females showing a faster rate of shortening than males (Supplementary Figure 1).

In an analysis exploring survival bias as an explanation for our results, we restricted testing to those younger than the median age (62 years). Multivariable linear regression accounting for sex and age still showed that telomeres were longer in people with ALS compared to controls (p = 8.12 × 10–12) with mean telomere length in people with ALS, 5.8 kb, and in controls, 5.5 kb. To ensure that telomere length analysis was not biased by population effects, we excluded the UK, a population we used previously for discovery analysis, and using a subset of samples the association was still observed (p = 6.6 × 10–9).

We compared telomere length in 334 people with ALS with C9orf72 repeat expansion against people with ALS with confirmed non-expanded C9orf72 status. Multivariable linear regression showed that the telomere was shorter in expansion carriers (p = 5.0 × 10–4) (Figure 2A and Table 3). Although ALS C9orf72 expansion carriers had a shorter telomere length than non-expansion carriers, telomere length was still longer in those carrying a C9orf72 repeat expansion than controls (p = 0.001) (Figure 2B).

FIGURE 2
www.frontiersin.org

Figure 2. (A) Telomere length comparison between 334 people with ALS carrying an expanded C9orf72 repeat against people with ALS not carrying an expansion. Multivariable linear regression shows that the telomere is shorter in those carrying a C9orf72 expansion compared with age and sex matched disease controls not carrying an expansion (p = 5.0 × 10– 4). (B) Telomere length comparison between 334 people with ALS with C9orf72 repeat expansion against 10 healthy individuals with confirmed expanded C9orf72 status. Multivariable linear regression shows that the telomere is longer in people with ALS with C9orf72 repeat expansion (p = 0.05). Telomere length reported in kilobases (kb). Red bars indicate 95% confidence intervals.

TABLE 3
www.frontiersin.org

Table 3. Telomere length comparison between 552 people with amyotrophic lateral sclerosis (ALS) with a C9orf72 repeat expansion and 907 people with ALS with normal C9orf72 repeat length using a multivariable linear regression.

We therefore assessed the relationship between the estimated number of telomere repeats and estimated number of C9orf72 repeats using ExpansionHunter in 1,589 samples from the UK. Multivariable linear regression showed that the number of telomere repeats is associated negatively with C9orf72 repeat expansion size (p = 0.003) supporting the previous results.

To assess if the relationship between C9orf72 repeat expansion and telomere repeat size was specific to C9orf72-mediated ALS, we ran ExpansionHunter on three ALS genes which also contain disease-associated repeat expansions: ATXN1, ATXN2, and NIPA1. There was no difference in telomere length observed between expansion carriers and non-expansion carriers for any of these genes.

Cox regression analysis showed that people with ALS with telomere length less than 5.3 Kb had a 10% increase in median survival compared with those with longer telomeres (p = 5.0 × 10–7) after correcting for age, sex, site of onset, C9orf72 status and principal components of ancestry.

To validate our findings in a different tissue we used 159 post-mortem brain samples, 106 from people with apparently sporadic ALS and 53 controls. The male-female ratio was 2:1. The mean telomere length in people with ALS was 6.8 kb, and in controls, 6.56 Kb, not taking into account gender or age. Multivariable linear regression accounting for these covariates showed that telomere length in people with ALS was longer by mean 29% (95% CI 30, 55%) compared with controls (p = 0.03).

Discussion

Using a large disease-specific whole genome sequencing dataset, we have shown that longer telomeres are associated with ALS, confirming initial findings from a pilot study (Al Khleifat et al., 2019). We were additionally able to show that our findings are likely a result of less rapid shortening of telomeres being associated with ALS, rather than active telomere lengthening in ALS. In keeping with expectations, we also found that mean telomere length was on average longer in females, and in all samples, shortened with increasing age. The association of longer telomeres with apparently sporadic ALS was also seen in FALS, supporting the notion that familial and sporadic ALS are not mutually exclusive categories but rather a spectrum (Al-Chalabi and Hardiman, 2013; Al-Chalabi et al., 2014; Chiò et al., 2018; Mehta et al., 2018). Furthermore, telomere length was inversely correlated with C9orf72 repeat expansion size.

Telomere elongation phenomena are well-documented but far less well-understood than telomere shortening phenomena (Bryan et al., 1995; Cesare and Reddel, 2010; Arora and Azzalin, 2015; Haycock et al., 2017). While telomere shortening is typically seen in cancers, telomere elongation can occur in cancers of the nervous system, and for example, is seen in 25% of primary brain tumors, in glioblastoma multiforme and in 10% of neuroblastomas (Bryan et al., 1997; Hakin-Smith et al., 2003; Henson et al., 2005; Durant, 2012; Boutou et al., 2013). In general, cancers in which cells have long telomeres are resistant to therapy and carry a poor prognosis (Haycock et al., 2017). Telomere elongation has also been associated with schizophrenia (Nieratschker et al., 2013; Zhang et al., 2018), a disorder that genetically overlaps with ALS (McLaughlin et al., 2017). Additionally, longer telomeres are also reported in Parkinson’s disease and Lewy body dementia blood and brain (Asghar et al., 2022).

We found that pathologically expanded C9orf72 repeats are negatively associated with telomere repeat length, so that people with expanded C9orf72 repeats had shorter telomeres on average. C9orf72 gene repeat expansion is the most frequent genetic cause of ALS and of the related condition, frontotemporal dementia (Shatunov et al., 2010; Smith et al., 2013; Hardiman et al., 2017; Iacoangeli et al., 2019). A possible explanation for the negative association is in the liability threshold model of disease. Those people who already have a high liability to ALS do not need the additional liability of longer telomeres and so on average would appear to have shorter telomeres. Alternatively, those in the higher risk group (non-repeat expansion carriers) need a greater contribution from other sources of disease liability and so have longer telomere repeats. Against this explanation is the observation that other gene variants that predispose to ALS risk such as intermediate ATXN2 repeat expansions, do not show any association with telomere length, and neither do people with familial ALS. The explanation might therefore lie in the C9orf72 repeat expansion itself. Both telomeres and large C9orf72 repeats have a tendency to fold into structures called G quadruplexes (Fratta et al., 2012; Grigg et al., 2014). G quadruplex structures have important roles in DNA replication, recombination and telomere maintenance (Millevoi et al., 2012; Bryan, 2019). Although there have been several studies of the G quadruplexes formed by C9orf72 repeat expansion, the relationship between C9orf72 repeats and other G quadruplexes such as telomeres is not documented at population level and has not been well-characterized in vivo (Zhang et al., 2019).

Some genetic variations that contribute to ALS risk also worsen prognosis. This is seen in carriers of the C9orf72 repeat expansion mutation, those carrying the UNC13A homozygous risk genotype, and for some variants of the SOD1 gene for example. In keeping with that pattern, we have found that longer telomere length is associated with ALS risk as well as with worse prognosis, with those with the shortest telomeres having a 10% increase in survival.

The genetic landscape of ALS is one of some monogenic causes, several gene variations that substantially but not dramatically increase risk, and a polygenic component. For those with a monogenic basis of their disease, the variable and age-dependent penetrance seen is likely because of the contribution of other factors to risk (Al-Chalabi and Hardiman, 2013; Al-Chalabi et al., 2014; Chiò et al., 2018; Garton et al., 2021). Based on these findings, telomere length could be such a factor.

The main limitation of this study is that we did not directly measure telomere length using Southern blotting, but estimated it using whole genome sequence data. The method we have used, TelSeq, was recently used to estimate telomere length in 75,000 whole genome sequences. Comparing the performance of TelSeq with other bioinformatics tools such as Computel, the estimates of telomere length were highly correlated between the bioinformatics methods and with Southern blot results, with the advantage that TelSeq had a faster processing time (Taub et al., 2019), although it is not possible to draw conclusions about the exact length of a telomere. Other studies have also shown a good correlation between TelSeq telomere length estimates, Southern blotting and Q-PCR (Ding et al., 2014; Cook et al., 2016). With this in mind, different sequencing technologies might generate different telomere length estimates because of differences in library preparation and platform (Ma, 1995; Aviv et al., 2011). To overcome this potential weakness, we have used the same industry-leading sequencing platform for all samples, as well as designing the study to minimize batch effects by having cases and controls sharing the same sequencing plate. Our study has the advantage of a large sample size of more than 4,500 cases, far larger than for previous reports.

We have shown that despite being an age-related, male-predominant condition, ALS is associated with longer telomere lengths in blood-derived and in brain-derived DNA.

Data availability statement

The data presented in this study are deposited in the Project MinE consortium public repository (http://databrowser.projectmine.com).

Ethics statement

The studies involving human participants were reviewed and approved by the Research Ethics Board at each respective recruiting site within the Project MinE consortium as previously described (van Rheenen et al., 2018).

Author contributions

AA-C and AAK conceived and planned the study, wrote the first draft of the manuscript, did the statistical analysis, and prepared the figures and tables. AAK and AI created the bioinformatics pipeline for analysis. JvV ran ExpansionHunter on Project MinE data. MM and RZ prepared phenotypic data. AA-C, JHV, OH, MP, JM, PS, JL, CS, NB, OH, WR, PVD, and LB helped in sample collection and provided whole genome sequence data and analysis and intellectual input for data interpretation on behalf of the Project MinE Consortium. JHV, OH, and MM provided intellectual input for data interpretation. All authors reviewed and approved the final manuscript.

Funding

AAK was funded by ALS Association Milton Safenowitz Research Fellowship (grant number 22-PDF-609. doi: 10.52546/pc.gr.150909), The Motor Neurone Disease Association (MNDA) Fellowship (Al Khleifat/Oct21/975-799), The Darby Rimmer Foundation, and The NIHR Maudsley Biomedical Research Centre. This project was also funded by the MND Association and the Wellcome Trust. This is an EU Joint Programme-Neurodegenerative Disease Research (JPND) project. The project is supported through the following funding organizations under the aegis of JPND–www.jpnd.eu [United Kingdom, Medical Research Council (MR/L501529/1 and MR/R024804/1) and Economic and Social Research Council (ES/L008238/1)]. AA-C was a NIHR Senior Investigator. CS and AA-C received salary support from the National Institute for Health Research (NIHR) Dementia Biomedical Research Unit at South London and Maudsley NHS Foundation Trust and King’s College London. The work leading up to this publication was funded by the European Community’s Health Seventh Framework Program (FP7/2007–2013; grant agreement number 259867) and Horizon 2020 Program (H2020-PHC-2014-two-stage; grant agreement number 633413). This project has received funding from the European Research Council (ERC) under the European Union’s Horizon 2020 Research and Innovation Programme (grant agreement no. 772376–EScORIAL. The collaboration project was co-funded by the PPP Allowance made available by Health∼Holland, Top Sector Life Sciences and Health, to stimulate public-private partnerships. Project MinE Belgium was supported by a grant from IWT, the Belgian ALS Liga and a grant from Opening the Future Fund (KU Leuven). PVD holds a senior clinical investigatorship of FWO-Vlaanderen and was supported by E. von Behring Chair for Neuromuscular and Neurodegenerative Disorders, the ALS Liga België and the KU Leuven funds “Een Hart voor ALS,” “Laeversfonds voor ALS Onderzoek,” and the “Valéry Perrier Race against ALS Fund”. RM was supported by Science Foundation Ireland (17/CDA/4737). MinE USA was funded by the US ALS Association.

Acknowledgments

Samples used in this research were in part obtained from the UK National DNA Bank for MND Research, funded by the MND Association and the Wellcome Trust. We thank people with MND and their families for their participation in this project. We acknowledge sample management undertaken by Biobanking Solutions funded by the Medical Research Council at the Centre for Integrated Genomic Medical Research, University of Manchester. The authors acknowledge use of the research computing facility at King’s College London, Rosalind (https://rosalind.kcl.ac.uk), which is delivered in partnership with the National Institute for Health Research (NIHR) Biomedical Research Centres at South London and Maudsley and Guy’s and St. Thomas’ NHS Foundation Trusts, and part-funded by capital equipment grants from the Maudsley Charity (award 980) and Guy’s and St. Thomas’ Charity (TR130505). We also acknowledge Health Data Research UK, which is funded by the UK Medical Research Council, Engineering and Physical Sciences Research Council, Economic and Social Research Council, Department of Health and Social Care (United Kingdom), Chief Scientist Office of the Scottish Government Health and Social Care Directorates, Health and Social Care Research and Development Division (Welsh Government), Public Health Agency (Northern Ireland), British Heart Foundation and Wellcome Trust.

Conflict of interest

AA-C was a consultant for Mitsubishi-Tanabe Pharma, GSK, and Chronos Therapeutics, and chief investigator for clinical trials for Cytokinetics and OrionPharma. JvV reports to have sponsored research agreements with Biogen. VS was a consultant for Novartis and Biogen. LB reports grants from Netherlands ALS Foundation, grants from Netherlands Organization for Health Research and Development (Vici Scheme), grants from The European Community’s Health Seventh Framework Programme [grant agreement no. 259867 (EuroMOTOR)], grants from Netherlands Organization for Health Research and Development) the STRENGTH project, funded through the EU Joint Programme—Neurodegenerative Disease Research, JPND), during the conduct of the study; personal fees from Calico, personal fees from Cytokinetics, grants and personal fees from Takeda, non-financial support from Orion, non-financial support from Orphazyme, outside the submitted work. AA-C also serves on scientific advisory boards for Mitsubishi Tanabe, Roche, Denali Pharma, Cytokinetics, Lilly, and Amylyx research. CS reports grants from Avexis, grants from Eli Lilly, grants from Chronos Therapeutics, grants from Vertex Pharmaceuticals, during the conduct of the study; grants from QurAlis, grants from Chronos Therapeutics, grants from Biogen, outside the submitted work. JL was a member of the scientific advisory board for Cerevel Therapeutics, a consultant for ACI Clinical LLC sponsored by Biogen, Inc. or Ionis Pharmaceuticals, Inc. JL was also a consultant for Perkins Coie LLP and may provide expert testimony and also supported by funding from NIH/NINDS (R01NS073873 and R56NS073873).

The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Author disclaimer

The views expressed are those of the authors and not necessarily those of the NHS, the NIHR or the Department of Health.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fncel.2022.1050596/full#supplementary-material

References

Al Khleifat, A., Iacoangeli, A., Shatunov, A., Fang, T., Sproviero, W., Jones, A. R., et al. (2019). Telomere length is greater in ALS than in controls: A whole genome sequencing study. Amyotroph. Lateral Scler. Frontotemporal Degener. 20, 229–234. doi: 10.1080/21678421.2019.1586951

PubMed Abstract | CrossRef Full Text | Google Scholar

Al-Chalabi, A., and Hardiman, O. (2013). The epidemiology of ALS: A conspiracy of genes, environment and time. Nat. Rev. Neurol. 9, 617–628. doi: 10.1038/nrneurol.2013.203

PubMed Abstract | CrossRef Full Text | Google Scholar

Al-Chalabi, A., Calvo, A., Chio, A., Colville, S., Ellis, C. M., Hardiman, O., et al. (2014). Analysis of amyotrophic lateral sclerosis as a multistep process: A population-based modelling study. Lancet Neurol. 13, 1108–1113. doi: 10.1016/S1474-4422(14)70219-4

CrossRef Full Text | Google Scholar

Al-Chalabi, A., Fang, F., Hanby, M. F., Leigh, P. N., Shaw, C. E., Ye, W., et al. (2010). An estimate of amyotrophic lateral sclerosis heritability using twin data. J. Neurol. Neurosurg. Psychiatry 81, 1324–1326. doi: 10.1136/jnnp.2010.207464

PubMed Abstract | CrossRef Full Text | Google Scholar

Arora, R., and Azzalin, C. M. (2015). Telomere elongation chooses TERRA ALTernatives. RNA Biol. 12, 938–941. doi: 10.1080/15476286.2015.1065374

PubMed Abstract | CrossRef Full Text | Google Scholar

Asghar, M., Odeh, A., Fattahi, A. J., Fattahi, A. J., Henriksson, A. E., Miglar, A., et al. (2022). Mitochondrial biogenesis, telomere length and cellular senescence in Parkinson’s disease and Lewy body dementia, scientific reports. Nat. Res. 12:17578. doi: 10.1038/s41598-022-22400-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Aviv, A., Hunt, S. C., Lin, J., Cao, X., Kimura, M., Blackburn, E., et al. (2011). Impartial comparative analysis of measurement of leukocyte telomere length/DNA content by Southern blots and qPCR. Nucleic Acids Res. 39:e134. doi: 10.1093/nar/gkr634

PubMed Abstract | CrossRef Full Text | Google Scholar

Boutou, E., Vlachodimitropoulos, D., Pappa, V., Stürzbecher, H.-T., and Vorgias, C. E. (2013). “DNA repair and telomeres — an intriguing relationship,” in New research directions in DNA repair, ed. C. Chen (Norderstedt: Books on Demand). doi: 10.5772/56115

CrossRef Full Text | Google Scholar

Brown, R. H., and Al-Chalabi, A. (2017). Amyotrophic lateral sclerosis. N. Engl. J. Med. 377, 162–172. doi: 10.1056/NEJMra1603471

PubMed Abstract | CrossRef Full Text | Google Scholar

Bryan, T. M. (2019). Mechanisms of DNA replication and repair: Insights from the study of G-quadruplexes. Molecules 24:3439. doi: 10.3390/molecules24193439

PubMed Abstract | CrossRef Full Text | Google Scholar

Bryan, T. M., Englezou, A., Dalla-Pozza, L., Dunham, M. A., and Reddel, R. R. (1997). Evidence for an alternative mechanism for maintaining telomere length in human tumors and tumor-derived cell lines. Nat. Med. 3, 1271–1274. doi: 10.1038/nm1197-1271

PubMed Abstract | CrossRef Full Text | Google Scholar

Bryan, T. M., Englezou, A., Gupta, J., Bacchetti, S., and Reddel, R. R. (1995). Telomere elongation in immortal human cells without detectable telomerase activity. EMBO J. 14, 4240–4248.

Google Scholar

Cesare, A. J., and Reddel, R. R. (2010). Alternative lengthening of telomeres: Models, mechanisms and implications. Nat. Rev. Genet. 11, 319–330. doi: 10.1038/nrg2763

PubMed Abstract | CrossRef Full Text | Google Scholar

Chiò, A., Mazzini, L., D’Alfonso, S., Corrado, L., Canosa, A., Moglia, C., et al. (2018). The multistep hypothesis of ALS revisited: The role of genetic mutations. Neurology 91, e635–e642. doi: 10.1212/WNL.0000000000005996

PubMed Abstract | CrossRef Full Text | Google Scholar

Conomos, D., Stutz, M. D., Hills, M., Neumann, A. A., Bryan, T. M., Reddel, R. R., et al. (2012). Variant repeats are interspersed throughout the telomeres and recruit nuclear receptors in ALT cells. J. Cell Biol. 199, 893–906. doi: 10.1083/jcb.201207189

PubMed Abstract | CrossRef Full Text | Google Scholar

Cook, D. E., Zdraljevic, S., Tanny, R. E., Seo, B., Riccardi, D. D., Noble, L. M., et al. (2016). The genetic basis of natural variation in Caenorhabditis elegans telomere length. Genetics 204, 371–383. doi: 10.1534/genetics.116.191148

PubMed Abstract | CrossRef Full Text | Google Scholar

Crockford, C., Newton, J., Lonergan, K., Chiwera, T., Booth, T., Chandran, S., et al. (2018). ALS-specific cognitive and behavior changes associated with advancing disease stage in ALS. Neurology 91, e1370–e1380. doi: 10.1212/WNL.0000000000006317

PubMed Abstract | CrossRef Full Text | Google Scholar

Ding, Z., Mangino, M., Aviv, A., Spector, T., and Durbin, R. (2014). Estimating telomere length from whole genome sequence data. Nucleic Acids Res. 42:e75. doi: 10.1093/nar/gku181

PubMed Abstract | CrossRef Full Text | Google Scholar

Dolzhenko, E., van Vugt, J. J. F. A., Shaw, R. J., Bekritsky, M. A., van Blitterswijk, M., Narzisi, G., et al. (2017). Detection of long repeat expansions from PCR-free whole-genome sequence data. Genome Res. 27, 1895–1903. doi: 10.1101/gr.225672.117

PubMed Abstract | CrossRef Full Text | Google Scholar

Durant, S. T. (2012). Telomerase-independent paths to immortality in predictable cancer sub-types. J. Cancer 3, 67–82. doi: 10.7150/jca.3965

PubMed Abstract | CrossRef Full Text | Google Scholar

Fratta, P., Mizielinska, S., Nicoll, A. J., Zloh, M., Fisher, E. M., Parkinson, G., et al. (2012). C9orf72 hexanucleotide repeat associated with amyotrophic lateral sclerosis and frontotemporal dementia forms RNA G-quadruplexes. Sci. Rep. 2:1016. doi: 10.1038/srep01016

PubMed Abstract | CrossRef Full Text | Google Scholar

Gardner, M., Bann, D., Wiley, L., Cooper, R., Hardy, R., Nitsch, D., et al. (2014). Gender and telomere length: Systematic review and meta-analysis. Exp. Gerontol. 51, 15–27. doi: 10.1016/j.exger.2013.12.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Garton, F. C., Trabjerg, B. B., Wray, N. R., and Agerbo, E. (2021). Cardiovascular disease, psychiatric diagnosis and sex differences in the multistep hypothesis of amyotrophic lateral sclerosis. Eur. J. Neurol. 28, 421–429. doi: 10.1111/ene.14554

PubMed Abstract | CrossRef Full Text | Google Scholar

Grigg, J. C., Shumayrikh, N., and Sen, D. (2014). G-quadruplex structures formed by expanded hexanucleotide repeat RNA and DNA from the neurodegenerative disease-linked C9orf72 gene efficiently sequester and activate heme. PLoS One 9:e106449. doi: 10.1371/journal.pone.0106449

PubMed Abstract | CrossRef Full Text | Google Scholar

Hakin-Smith, V., Jellinek, D. A., Levy, D., Carroll, T., Teo, M., Timperley, W. R., et al. (2003). Alternative lengthening of telomeres and survival in patients with glioblastoma multiforme. Lancet 361, 836–838. doi: 10.1016/S0140-6736(03)12681-5

CrossRef Full Text | Google Scholar

Hardiman, O., Al-Chalabi, A., Chio, A., Corr, E. M., Logroscino, G., Robberecht, W., et al. (2017). Amyotrophic lateral sclerosis. Nat. Rev. Dis. Primers 3:17071. doi: 10.1038/nrdp.2017.71

PubMed Abstract | CrossRef Full Text | Google Scholar

Haycock, P. C., Burgess, S., Nounu, A., Zheng, J., Okoli, G. N., Bowden, J., et al. (2017). Association between telomere length and risk of cancer and non-neoplastic diseases a mendelian randomization study. JAMA Oncol. 3, 636–651. doi: 10.1001/jamaoncol.2016.5945

PubMed Abstract | CrossRef Full Text | Google Scholar

Henson, J. D., Hannay, J. A., McCarthy, S. W., Royds, J. A., Yeager, T. R., Robinson, R. A., et al. (2005). A robust assay for alternative lengthening of telomeres in tumors shows the significance of alternative lengthening of telomeres in sarcomas and astrocytomas. Clin. Cancer Res. 11, 217–225.

PubMed Abstract | Google Scholar

Hop, P. J., Zwamborn, R. A., Hannon, E., Shireby, G. L., Nabais, M. F., Walker, E. M., et al. (2022). Genome-wide study of DNA methylation in amyotrophic lateral sclerosis identifies differentially methylated loci and implicates metabolic, inflammatory and cholesterol pathways. Philippe Couratier 18:37. doi: 10.1101/2021.03.12.21253115

CrossRef Full Text | Google Scholar

Iacoangeli, A., Al Khleifat, A., Jones, A. R., Sproviero, W., Shatunov, A., Opie-Martin, S., et al. (2019). C9orf72 intermediate expansions of 24–30 repeats are associated with ALS. Acta Neuropathol. Commun. 7:115. doi: 10.1186/s40478-019-0724-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Kong, C. M., Lee, X. W., and Wang, X. (2013). Telomere shortening in human diseases. FEBS J. 280, 3180–3193. doi: 10.1111/febs.12326

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, T. S. (1995). Applications and limitations of polymerase chain reaction amplification. Chest 108, 1393–1404. doi: 10.1378/chest.108.5.1393

PubMed Abstract | CrossRef Full Text | Google Scholar

Marzec, P., Armenise, C., Pérot, G., Roumelioti, F. M., Basyuk, E., Gagos, S., et al. (2015). Nuclear-receptor-mediated telomere insertion leads to genome instability in ALT cancers. Cell 160, 913–927. doi: 10.1016/j.cell.2015.01.044

PubMed Abstract | CrossRef Full Text | Google Scholar

McCombe, P. A., and Henderson, R. D. (2010). Effects of gender in amyotrophic lateral sclerosis. Gend. Med. 7, 557–570. doi: 10.1016/j.genm.2010.11.010

PubMed Abstract | CrossRef Full Text | Google Scholar

McLaughlin, L. R., Vajda, A., and Hardiman, O. (2015). Heritability of amyotrophic lateral sclerosis insights from disparate numbers. JAMA Neurol. 72, 857–858. doi: 10.1001/jamaneurol.2014.4049

PubMed Abstract | CrossRef Full Text | Google Scholar

McLaughlin, R. L., Schijven, D., van Rheenen, W., van Eijk, K. R., O’Brien, M., Kahn, R. S., et al. (2017). Genetic correlation between amyotrophic lateral sclerosis and schizophrenia. Nat. Commun. 8:14774. doi: 10.1038/ncomms14774

PubMed Abstract | CrossRef Full Text | Google Scholar

Mehta, P. R., Jones, A. R., Opie-Martin, S., Shatunov, A., Iacoangeli, A., Khleifat, A., et al. (2018). Younger age of onset in familial amyotrophic lateral sclerosis is a result of pathogenic gene variants, rather than ascertainment bias. J. Neurol. Neurosurg. Psychiatry 90, 268–271. doi: 10.1136/jnnp-2018-319089

PubMed Abstract | CrossRef Full Text | Google Scholar

Millevoi, S., Moine, H., and Vagner, S. (2012). G-quadruplexes in RNA biology. Wiley Interdiscip. Rev. RNA 3, 495–507. doi: 10.1002/wrna.1113

PubMed Abstract | CrossRef Full Text | Google Scholar

Murnane, J. P. (2006). Telomeres and chromosome instability. DNA Repair 5, 1082–1092. doi: 10.1016/j.dnarep.2006.05.030

PubMed Abstract | CrossRef Full Text | Google Scholar

Muzumdar, R., and Atzmon, G. (2012). Telomere length and aging. Rev. Sel. Top. Telomere Biol. 1, 3–30. doi: 10.5772/2329

CrossRef Full Text | Google Scholar

Nieratschker, V., Lahtinen, J., Meier, S., Strohmaier, J., Frank, J., Heinrich, A., et al. (2013). Longer telomere length in patients with schizophrenia. Schizophr. Res. 149, 116–120. doi: 10.1016/j.schres.2013.06.043

PubMed Abstract | CrossRef Full Text | Google Scholar

Raczy, C., Petrovski, R., Saunders, C. T., Chorny, I., Kruglyak, S., Margulies, E. H., et al. (2013). Isaac: Ultra-fast whole-genome secondary analysis on illumina sequencing platforms. Bioinformatics 29, 2041–2043. doi: 10.1093/bioinformatics/btt314

PubMed Abstract | CrossRef Full Text | Google Scholar

Roche, J. C., Rojas-Garcia, R., Scott, K. M., Scotton, W., Ellis, C. E., Burman, R., et al. (2012). A proposed staging system for amyotrophic lateral sclerosis. Brain 135, 847–852. doi: 10.1093/brain/awr351

PubMed Abstract | CrossRef Full Text | Google Scholar

Shatunov, A., Mok, K., Newhouse, S., Weale, M. E., Smith, B., Vance, C., et al. (2010). Chromosome 9p21 in sporadic amyotrophic lateral sclerosis in the UK and seven other countries: A genome-wide association study. Lancet Neurol. 9, 986–994. doi: 10.1016/S1474-4422(10)70197-6

CrossRef Full Text | Google Scholar

Smith, B. N., Newhouse, S., Shatunov, A., Vance, C., Topp, S., Johnson, L., et al. (2013). The C9ORF72 expansion mutation is a common cause of ALS+/-FTD in Europe and has a single founder. Eur. J. Hum. Genet. 21, 102–108. doi: 10.1038/ejhg.2012.98

PubMed Abstract | CrossRef Full Text | Google Scholar

Taub, M. A., Conomos, M. P., Keener, R., Iyer, K. R., Weinstock, J. S., Yanek, L. R., et al. (2019). Novel genetic determinants of telomere length from a multi-ethnic analysis of 75,000 whole genome sequences in TOPMed. bioRxiv [Preprint]. doi: 10.1101/749010

CrossRef Full Text | Google Scholar

van Rheenen, W., Sara, L. P., Annelot, M. D., Al Khleifat, A., William, J. B., Kevin, P., et al. (2018). Project MinE: Study design and pilot analyses of a large-scale whole-genome sequencing study in amyotrophic lateral sclerosis. Eur. J. Hum. Genet. 26, 1537–1546. doi: 10.1038/s41431-018-0177-4

PubMed Abstract | CrossRef Full Text | Google Scholar

van Rheenen, W., Shatunov, A., Dekker, A. M., McLaughlin, R. L., Diekstra, F. P., Pulit, S. L., et al. (2016). Genome-wide association analyses identify new risk variants and the genetic architecture of amyotrophic lateral sclerosis. Nat. Genet. 48, 1043–1048. doi: 10.1038/ng.3622

PubMed Abstract | CrossRef Full Text | Google Scholar

van Rheenen, W., van der Spek, R. A. A., Bakker, M. K., van Vugt, J. J. F. A., Hop, P. J., Zwamborn, R. A. J., et al. (2021). Common and rare variant association analyses in amyotrophic lateral sclerosis identify 15 risk loci with distinct genetic architectures and neuron-specific biology. Anneke J. Van Der Kooi 17:129. doi: 10.1101/2021.03.12.21253159

CrossRef Full Text | Google Scholar

Westeneng, H. J., Debray, T. P. A., Visser, A. E., van Eijk, R. P. A., Rooney, J. P. K., Calvo, A., et al. (2018). Prognosis for patients with amyotrophic lateral sclerosis: Development and validation of a personalised prediction model. Lancet Neurol. 17, 423–433. doi: 10.1016/S1474-4422(18)30089-9

CrossRef Full Text | Google Scholar

Xu, L., Li, S., and Stohr, B. A. (2013). The role of telomere biology in cancer. Annu. Rev. Pathol. 8, 49–78. doi: 10.1146/annurev-pathol-020712-164030

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, S., Sun, H., Yang, D., Liu, Y., Zhang, X., Chen, H., et al. (2019). Evaluation of the selectivity of G-quadruplex ligands in living cells with a small molecule fluorescent probe. Anal. Chim. Acta X 2:100017. doi: 10.1016/j.acax.2019.100017

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Hishimoto, A., Otsuka, I., Watanabe, Y., Numata, S., Yamamori, H., et al. (2018). Longer telomeres in elderly schizophrenia are associated with long-term hospitalization in the Japanese population. J. Psychiatr. Res. 103, 161–166. doi: 10.1016/j.jpsychires.2018.05.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: amyotrophic lateral sclerosis (ALS), telomere–genetics, whole genome sequence (WGS), genomics, bigdata, MND–motor neuron disorders

Citation: Al Khleifat A, Iacoangeli A, Jones AR, van Vugt JJFA, Moisse M, Shatunov A, Zwamborn RAJ, van der Spek RAA, Cooper-Knock J, Topp S, van Rheenen W, Kenna B, Van Eijk KR, Kenna K, Byrne R, López V, Opie-Martin S, Vural A, Campos Y, Weber M, Smith B, Fogh I, Silani V, Morrison KE, Dobson R, van Es MA, McLaughlin RL, Vourc’h P, Chio A, Corcia P, de Carvalho M, Gotkine M, Panades MP, Mora JS, Shaw PJ, Landers JE, Glass JD, Shaw CE, Basak N, Hardiman O, Robberecht W, Van Damme P, van den Berg LH, Veldink JH and Al-Chalabi A (2022) Telomere length analysis in amyotrophic lateral sclerosis using large-scale whole genome sequence data. Front. Cell. Neurosci. 16:1050596. doi: 10.3389/fncel.2022.1050596

Received: 21 September 2022; Accepted: 15 November 2022;
Published: 15 December 2022.

Edited by:

Agnes Lumi Nishimura, Queen Mary University of London, United Kingdom

Reviewed by:

Danyllo Oliveira, University of São Paulo, Brazil
Christos Proukakis, University College London, United Kingdom

Copyright © 2022 Al Khleifat, Iacoangeli, Jones, van Vugt, Moisse, Shatunov, Zwamborn, van der Spek, Cooper-Knock, Topp, van Rheenen, Kenna, Van Eijk, Kenna, Byrne, López, Opie-Martin, Vural, Campos, Weber, Smith, Fogh, Silani, Morrison, Dobson, van Es, McLaughlin, Vourc’h, Chio, Corcia, de Carvalho, Gotkine, Panades, Mora, Shaw, Landers, Glass, Shaw, Basak, Hardiman, Robberecht, Van Damme, van den Berg, Veldink and Al-Chalabi. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Ammar Al-Chalabi, ammar.al-chalabi@kcl.ac.uk; Ahmad Al Khleifat, ahmad.al_khleifat@kcl.ac.uk

These authors have contributed equally to this work

Download