+ All Categories
Home > Documents > Stem Cell Research, 30: 150-162 Citation for the or iginal...

Stem Cell Research, 30: 150-162 Citation for the or iginal...

Date post: 11-Aug-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
14
http://www.diva-portal.org This is the published version of a paper published in Stem Cell Research. Citation for the original published paper (version of record): Higelin, J., Catanese, A., Semelink-Sedlacek, L L., Oeztuerk, S., Lutz, A-K. et al. (2018) NEK1 loss-of-function mutation induces DNA damage accumulation in ALS patient- derived motoneurons Stem Cell Research, 30: 150-162 https://doi.org/10.1016/j.scr.2018.06.005 Access to the published version may require subscription. N.B. When citing this work, cite the original published paper. Permanent link to this version: http://urn.kb.se/resolve?urn=urn:nbn:se:umu:diva-150372
Transcript
Page 1: Stem Cell Research, 30: 150-162 Citation for the or iginal ...umu.diva-portal.org/smash/get/diva2:1237422/FULLTEXT01.pdf · This is the published version of a paper published in Stem

http://www.diva-portal.org

This is the published version of a paper published in Stem Cell Research.

Citation for the original published paper (version of record):

Higelin, J., Catanese, A., Semelink-Sedlacek, L L., Oeztuerk, S., Lutz, A-K. et al. (2018)NEK1 loss-of-function mutation induces DNA damage accumulation in ALS patient-derived motoneuronsStem Cell Research, 30: 150-162https://doi.org/10.1016/j.scr.2018.06.005

Access to the published version may require subscription.

N.B. When citing this work, cite the original published paper.

Permanent link to this version:http://urn.kb.se/resolve?urn=urn:nbn:se:umu:diva-150372

Page 2: Stem Cell Research, 30: 150-162 Citation for the or iginal ...umu.diva-portal.org/smash/get/diva2:1237422/FULLTEXT01.pdf · This is the published version of a paper published in Stem

Contents lists available at ScienceDirect

Stem Cell Research

journal homepage: www.elsevier.com/locate/scr

NEK1 loss-of-function mutation induces DNA damage accumulation in ALSpatient-derived motoneurons

Julia Higelina,b,1, Alberto Catanesea,b,2, Lena Luisa Semelink-Sedlaceka, Sertap Oeztuerka,Anne-Kathrin Lutza,b, Julia Bausingerc, Gotthold Barbic, Günter Speitc, Peter M. Andersend,Albert C. Ludolphe, Maria Demestrea,⁎, Tobias M. Boeckersa,⁎

a Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germanyb International Graduate School in Molecular Medicine, Ulm University, Ulm, Germanyc Institute for Human Genetics, University Ulm, Ulm, Germanyd Department of Pharmacology and Clinical Neuroscience, Umea University, Umea, Swedene Department of Neurology, University Ulm, Ulm, Germany

A R T I C L E I N F O

Keywords:hiPSCALSNEK1NeurodegenerationDNA damage

A B S T R A C T

Mutations in genes coding for proteins involved in DNA damage response (DDR) and repair, such as C9orf72 andFUS (Fused in Sarcoma), are associated with neurodegenerative diseases and lead to amyotrophic lateralsclerosis (ALS). Heterozygous loss-of-function mutations in NEK1 (NIMA-related kinase 1) have also been re-cently found to cause ALS. NEK1 codes for a multifunctional protein, crucially involved in mitotic checkpointcontrol and DDR. To resolve pathological alterations associated with NEK1 mutation, we compared hiPSC-de-rived motoneurons carrying a NEK1mutation with mutant C9orf72 and wild type neurons at basal level and afterDNA damage induction. Motoneurons carrying a C9orf72 mutation exhibited cell specific signs of increased DNAdamage. This phenotype was even more severe in NEK1c.2434A>T neurons that showed significantly increasedDNA damage at basal level and impaired DDR after induction of DNA damage in an maturation-dependentmanner. Our results provide first mechanistic insight in pathophysiological alterations induced by NEK1 mu-tations and point to a converging pathomechanism of different gene mutations causative for ALS. Therefore, ourstudy contributes to the development of novel therapeutic strategies to reduce DNA damage accumulation inneurodegenerative diseases and ALS.

1. Introduction

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative diseasecharacterized by the selective death of upper and lower motoneurons(MN), which leads to muscle denervation and eventually death by re-spiratory failure (Boillée et al., 2006). Although most ALS cases do nothave a familial history (sporadic cases), specific mutations in differentgenes could be identified in about 10% of ALS patients (familial cases,i.e. fALS). In the last decades, several new genes have been identifiedthat are associated with familial and sporadic ALS (Chen et al., 2013;

Taylor et al., 2016). The complex pathogenic landscape and the func-tional heterogeneity of ALS genes are limiting factors for the develop-ment of effective ALS therapies. In fact, although eventually leading tothe same disease phenotype, ALS-associated genetic mutations occur ingenes that are involved in a plethora of different cellular pathways suchas protein degradation, RNA processing, and DNA-damage repair(Lagier-Tourenne et al., 2010; Qiu et al., 2014; Vance et al., 2009; Wanget al., 2013).

Recently, whole genome sequencing of ALS patients and healthyindividuals revealed an association of heterozygous mutations in the

https://doi.org/10.1016/j.scr.2018.06.005Received 29 January 2018; Received in revised form 22 May 2018; Accepted 7 June 2018

⁎ Corresponding authors at: Institute of Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, Ulm 89073, Germany.

1 These authors contributed equally to this work.2 These authors contributed equally to this work.

E-mail addresses: [email protected] (M. Demestre), [email protected] (T.M. Boeckers).

Abbreviations: ALS, amyotrophic lateral sclerosis; ATM (serine/threonine kinase), BRCA1, Breast Cancer 1; CHK1, Checkpoint kinase 1; DDR, DNA damage response; DSB, doublestrand break; hiPSC, human induced pluripotent stem cells; HR, homologous recombination; NEK1, NIMA-related kinase 1; NHEJ, non-homologous end joining

Stem Cell Research 30 (2018) 150–162

Available online 12 June 20181873-5061/ © 2018 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/BY-NC-ND/4.0/).

T

Page 3: Stem Cell Research, 30: 150-162 Citation for the or iginal ...umu.diva-portal.org/smash/get/diva2:1237422/FULLTEXT01.pdf · This is the published version of a paper published in Stem

mitotic protein kinase never-in-mitosis A (NIMA-) related kinase 1(NEK1) with ALS (Brenner et al., 2016; Cirulli et al., 2015). Thus, thesestudies highlighted NEK1 as a novel ALS gene. NEK1 is known to beinvolved in DNA damage response (DDR), since it specifically localizesto DNA damage foci after γ-irradiation (Chen et al., 2008; Polci et al.,2004). Moreover, cells lacking NEK1 display severe alterations in cellcycle checkpoints control, since the protein is required for activation ofthe cell cycle kinases CHK1 and CHK2 (checkpoint kinase 1/2). Bothkinases are indispensable for proper arrest at G1/S, S- or G2/M-phaseand efficient DNA repair after DNA damage (Chen et al., 2008). Con-sequently, NEK1-deficient cells suffer from unrepaired DNA damage,finally leading to chromosomal breaks and genomic instability (Chenet al., 2011).

Amongst others, ALS-related mutations have also been shown toaffect different genes specifically involved in the maintenance ofgenomic stability and DNA repair mechanisms. For instance, mutationsin Fused-in-Sarcoma (FUS) (Qiu et al., 2014; Wang et al., 2013) andC9orf72 (Lopez-Gonzalez et al., 2016) are associated with ALS and ithas already been shown that they are causative of increased DNA da-mage. However, it is not known whether an ALS related NEK1 mutationcauses increased DNA damage and impaired DDR after induction ofDNA strand breaks. Since NEK1 is a key element in early DNA damagerepair pathways (Chen et al., 2008, 2011; Polci et al., 2004) and alteredDDR has already been associated with fatal neurodegeneration and ALSprogression, we analyzed and characterized iPSC-derived MN derivedfrom a newly generated NEK1c.2434A>T cell line. Based on this, weespecially focused on the question whether the NEK1 mutation is in-deed leading to haploinsufficiency causing alterations in DDR as al-ready been demonstrated for C9orf72 (Lopez-Gonzalez et al., 2016).The analysis aims to clarify if mutations in different ALS related genescould lead to the same ALS associated motoneuronal pathology even-tually leading to cell death.

2. Material and methods

2.1. Cultivation of human keratinocytes

Cultivation of keratinocytes from plucked human hair and cultiva-tion CD-1 mouse embryonic fibroblasts (MEF) (Stemcell Technologies)was performed as already described (Aasen et al., 2008; Takahashi andYamanaka, 2006). Briefly, keratinocytes obtained from the out-growthof several hair roots were cultured in EpiLife medium with HKGSsupplement (Invitrogen) on collagen IV-coated (20 μg/ml; Sigma-Al-drich) dishes. MEFs were cultivated according to manufacturer's pro-tocol.

2.2. Generation of human iPS cells and characterization of pluripotency

Generation of human iPS cells by reprogramming human keratino-cytes was performed as previously described (Stockmann et al., 2013).Lentiviral particles were produced as already published (Linta et al.,2012; Sommer et al., 2016; Stockmann et al., 2013; Warlich et al.,2011). Briefly, 3–105 cells were infected in EpiLife medium containing8mg/ml polybrene (Sigma-Alrich). Infection was performed over 2successive days. After expansion, iPSC colonies were transferred ontoMatrigel-coated (BD Biosciences) plates. Pluripotency tests were doneusing the StemLite Pluripotency Kit (Cell Signaling). In vitro differ-entiation necessary for germ-layer-specific verification was performedaccording to published protocols (Linta et al., 2012; Stockmann et al.,2013). Chromosome preparation for Karyotyping was carried out

according to standard protocols (Linta et al., 2012) (supplemental dataFig. S1D).

2.3. Sequencing of NEK1c.2434A>T and CNTL2

To verify the presence of described mutation c.2434A > T withinthe NEK1 gene, affected area was sequenced for cell line NEK1c.2434A>T

and CNTL2For that, total DNA was isolated using the QIAamp DNA Mini Kit

(Qiagen) according to manufacturers` protocol. A PCR using a specificprimer pair (CTGATACCCGGGAAACTTCA, TGTGAGAGGGAGGCACTTCT) flanking the mutation site was performed to obtain a fragment,suitable for sequencing. After separation via Electrophoresis respectiveband (521 bp) was isolated using Gel Extraction Spin Kit 150(Genomed) according to manufacturers` protocol. All samples were sentand sequenced by mwg-biotech.

2.4. Cultivation of human iPS cells and motoneuronal differentiation

Cultivation of reprogrammed hiPSCs under feeder-free conditions aswell the differentiation of hiPSCs into mature spinal MNs was per-formed as previously described (Hu and Zhang, 2009; Stockmann et al.,2013). Differentiated cells were cultured on 35mm dishes (Ibidi) andkept in culture for up to 42 days.

2.5. Semi-quantitative real-time one-step RT-PCR

Semi-quantitative real-time one-step RT-PCR was performed as de-scribed (Kleger et al., 2010) using a Rotor-Gene Q System (Qiagen). 3technical replicates of each cell line (n=3) were used and results areshown relative to housekeeping gene HMBS. Validated primers pairswere purchased from Qiagen (Quantitect primer assay, Qiagen).

2.6. Fluorescence in situ hybridization (FISH)

To perform fluorescence in situ hybridization (FISH) cellswere washed with phosphate buffer saline (PBS), fixed 4%Paraformaldelhyde (PFA) and permeabilized with 0.2% Triton X-100(Roche). The Cy3-labelled probe against hexanucleotide repeatGGGGCC was dissolved in 50% formamide in 2× saline‑sodium citrate(SSC) buffer plus 10% dextran sulphate (probe concentration 0.02 ng/μl). To hinder unspecific binding, 1 μg/ μl of salmon sperm DNA wasadded. The probe was denatured for 5min at 60 °C, then kept for 3minon ice. Next, the probe was added to the cells and incubated for 3-4 h at42 °C in a humid, dark chamber. After incubation, cells were washedwith preheated solutions (42 °C): three times 5min with 50% for-mamide in 2× SSC, three times 5min with 2× SSC and once for 5minwith 4× SSC plus 0.03% Tween 20 at RT. Then, cells were washed withPBS and aqua demin. Finally, cover slips were mounted with ProLongGold Antifade reagent with Dapi (Invitrogen).

2.7. Southern Blot

Southern Blot was performed according to a standard protocol de-scribed in previously published manuscripts (Hübers et al., 2014).Briefly, enzymatically digested DNA fragments were separated using aTRIS-Borat-EDTA agarose gel (0.9%), transferred onto Amersham Hy-bond NTM-XL (Fisher Scientific) and incubated with a 32P-labeld probe.

J. Higelin et al. Stem Cell Research 30 (2018) 150–162

151

Page 4: Stem Cell Research, 30: 150-162 Citation for the or iginal ...umu.diva-portal.org/smash/get/diva2:1237422/FULLTEXT01.pdf · This is the published version of a paper published in Stem

2.8. Western Blot

All Western Blots were carried out as previously described(Grabrucker et al., 2011) using 3 technical replicates of each cell line(n=3). Briefly, total protein concentration of whole cell lysates wasdefined by Bradford Assay and equal protein amounts were separatedthrough a polyacrylamide gel electrophoresis (SDS-PAGE). Subse-quently, separated samples were transferred on a nitrocellulose mem-brane (GE Healthcare). Primary antibodies (NEK1, mouse, 1:500, santacruz; Caspase-3, rabbit, 1:500, cell signaling; pBRCA1, rabbit, 1 μg/ml,abcam; BRCA1, rabbit, 1:500, abcam; Ku70, 1:1000, rabbit abcam;ATR; pATR, both rabbit, 1:1000, cell signaling) were detected usinghorseradish peroxidase- (HRP) conjugated secondary antibodies andthe ECL detection Kit (Thermo Scientific). Primary Anti-NEK1 was de-tected using m-IgGК BP-HRP (mouse, 1:3000, santa cruz). Gel-analyzerSofware2 was used for quantification. Obtained values were normalizedagainst load control β-actin (1:250.000, Sigma Aldrich) and presentedrelative to untreated CNTL1.

2.9. Immunocytochemistry

Immunofluorescence was performed as described in standard pro-tocols (Stockmann et al., 2013). Cells were fixed with 4% PFA/PBS andprimary antibodies were directed against: ChAT (rabbit, 1:500, abcam),NEK1 (rabbit, 1:1000, abcam), NF-H (chicken, 1:50.000, Antibodiesonline), γH2A.X phosphor S139 (mouse, 1:2000, abcam), Actinin(mouse, 1:200, chemcon), β-Catenin (mouse, 1:500, abcam) Tubulin-β-III (chicken, 1:1000, merck) and incubated for 12 h at 4 °C. PrimaryAntibodies were detected with fluorescence labelled secondary anti-bodies (all 1:500, Invitrogen): Alexa Fluor® 488, Alexa Fluor® 568,Alexa Fluor® 647 and mounted with ProLong Gold Antifade reagent.For quantification analysis, 3 technical replicates of each line wereanalyzed (n=3).

2.10. Induction of DNA damage by γ-irradiation

DNA damage was induced by γ-irradiation (0.5 Gy) in un-differentiated hiPSCs as well as in MNs (21 days, 42 days). HiPS cellswere lifted via hESC-dispase digestion (Stemcell Technologies), seededon hESC-qualified-matrigel coated 35mm dishes (Ibidi) and culturedfor 3–5 days. Medium was changed 1 h before colonies were exposed toγ-irradiation. IPSCs were then cultured in mTeSR1 medium (StemcellTechnologie) for 1 h/24 h after treatment and either fixed for im-munochemical analysis or prepared for Western blot analysis. To ana-lyze the effect of irradiation in MNs, neuronal spheres were plated on35mm dishes. Cells were kept in culture for 21/42 days with changingmedium 2 times per week. Medium was changed 1 h before cultureswere treated. After 1 h/24 h of irradiation, cells were either fixed in 4%PFA/PBS or prepared for Western blot analysis.

2.11. Comet assay

Comet Assay was performed according to a standard protocol (Speitand Rothfuss, 2012). After corresponding treatment (without/1 h/24 hafter irradiation), cells were lifted via Accutase (Sigma Aldrich) for3min at 37 °C. Afterwards, to ensure single-cell suspension sampleswere filtered through 30 μm Pre-Separation filters (Miltenyi Biotec).15,000 cells per sample were mixed with 120 μl low melting-pointagarose (0.5% in PBS) and added to microscope slides which had beencovered with a bottom layer of 1.5% agarose. Slides were lysed for atleast 1 h at 4 °C (pH 10) and alkali denaturated for 25min (pH > 13).Electrophoresis was performed at 0.86 V/cm for 25min. Slides were

coded and images of 100 randomly selected cells stained with ethidiumbromide were analyzed from each slide. Values were calculated usingimage analysis (Comet Assay IV, Perceptive Instruments, Haverhill).DNA migration was calculated by analyzing “tail length” and “tail in-tensity (% tail DNA)” as a mean of three experiments (3 technical re-plicates for each cell line), which were independently performed underthe same experimental conditions.

2.12. Caspase-3 and ROS assay

Caspase-3 activity in 42 days old MNs ± irradiation was done ac-cording to the manufacturer's protocol using the colorimetric Caspase 3Assay Kit (abcam). Identification of the amount of reactive oxygenspecies (ROS) was performed as described in the manual of OxiSelectIntracellular ROS Assay Kit (Green Fluorescence) (Cell Biolabs Inc.). Foranalysis, 3 technical replicates were used f cell line.

2.13. Cell cycle phospho antibody array kit detection and analysis

The phospho antibody array kit (Full Moon Biosystems) was per-formed according to the manufacturer's protocol using 3 technical re-plicates for each line (n=3). Briefly, MNs were lysated, centrifugedand stored at −80 °C until analysis. 25 μl lysate containing 39 μg pro-tein was mixed with 75 μl labeling buffer (provided by manufacturer)and biotinylated for 2 h at RT. Biotinylated samples were stored at−80 °C. Blocking and coupling was performed according to the man-ufacturer's protocol. The detection of antibody arrays was performed ina fluorescent slide scanner (Genepix 4000B microarray scanner,Molecular Devices). The 16-bits images were analyzed using GenePixPro 6.1 software. For quantification, 6 replicates for each antibodydetectio and values of three independent experiments were used. Valueswere calculated as relative to the average value of correspondingCNTRL.

2.14. Statistical analysis

Fluorescence images were captured with an upright Axioscope mi-croscope provided with a CCD camera (16 bits; 1280×1024 pixels)using the software Axiovision (Zeiss). For Quantification analysis(γH2A.X+ cells), 4–9 representative images were taken randomly fromrespective conditions (± irradiation) and at least 200 cells were ana-lyzed for each condition using the Image J Fiji Software (www.imageJ.nih.gov) (Schindelin et al., 2012). Each experiment was repeated atleast 3 times. Statistical analysis of all data was performed usingGraphPad Prism 5 (GraphPad Software Inc., La Jolla, CA) software.Results are represented as mean values± standard error of the mean(SEM). Significance is shown as * p≤ .05, ** p≤ .01, and *** p≤ .001.Statistically significant differences were determined according to theparameters either by two-way ANOVA with Bonferroni post-test, one-way ANOVA with Bonferroni post-test or unpaired t-test. Statisticalanalysis of DNA damage categorization was performed using two-wayrepeated measures ANOVA which accounted for the measurement tri-plets of our experiments. Counts were considered as percentages ratherthan absolute numbers. A Bonferroni adjustment was applied to ac-count for the pairwise multiple comparisons of different cell lines.

2.15. Ethical statement

Experiments with human material were in accordance with theethical committee of Ulm University (Nr.0148/2009; 265/12) and incompliance with guidelines of the Federal Government of Germany (Nr.O.103).

J. Higelin et al. Stem Cell Research 30 (2018) 150–162

152

Page 5: Stem Cell Research, 30: 150-162 Citation for the or iginal ...umu.diva-portal.org/smash/get/diva2:1237422/FULLTEXT01.pdf · This is the published version of a paper published in Stem

Key resources table

Reagent or resource Source Identifier

AntibodiesMouse monoclonal anti Nek1 Santa cruz sc-398813Rabbit polyclonal anti Nek1 abcam ab186519Rabbit polyclonal anti pBRCA1 abcam ab119381Rabbit polyclonal anti BRCA1 abcam ab9141Rabbit polyclonal anti Ku70 abcam ab83501Rabbit anti ATR Cell signalling Cat. #2790Rabbit anti pATR (Thr1989) Cell signalin Cat. #58014Rabbit anti Caspase-3 Cell signaling Cat. #9662Rabbit polyclonal anti ChAT abcam ab18736Chicken polyclonal anti NF-H Antibodies online ABIN1842223Mouse monoclonal anti gamma H2A.X

(phospho S139)abcam ab26350

Mouse monoclonal anti actinin sigma A7811Mouse monoclonal anti AFP R&D systems MAB1368Mouse monoclonal ß-Catenin abcam ab22656Chicken polyclonal anti Tubulin-ß-III merck AB9354Polyclonal Alexa Fluor® 488 goat anti rabbit Invitrogen A-11034Polyclonal Alexa Fluor® 568 goat anti mouse Invitrogen A-11004Polyclonal Alexa Fluor® 647 goat anti chicken Invitrogen A21449m-IgGκ BP-HRP Santa cruz sc-516102

Bacterial and Virus StrainsLentivirus (polycistronic expression cassette) produced with Lenti-X 293T cells as described in (Sommer et al., 2016) N/A

Biological SamplesHuman hair sample CNTL1 RKU Uni-Ulm

Oberer Eselsberg 4589081 Ulm

N/A

Human hair sample CNTL2 Clinical Neuroscience, Umeå UniversitySE-90185 Umeå

N/A

Human hair sample C9ORF RKU Uni-UlmOberer Eselsberg 4589081 Ulm

N/A

Human hair sample NEK1 Clinical Neuroscience, Umeå UniversitySE-90185 Umeå

N/A

Chemicals, Peptides, and Recombinant Proteins

Critical Commercial Assayscolorimetric Caspase 3 Assay Kit abcam ab39401OxiSelect Intracellular ROS Assay Kit Cell Biolabs Inc STA-342-5Comet assay (Speit & Rothfuss, 2012) N/AStemLite Pluripotency Kit Cell Signaling Cat.#9656Cell Cycle Phospho Antibody Array Full Moon Biosystems Cat.#PCC076

Deposited Data

Experimental Models: Cell LinesCD-1 mouse embryonic fibroblasts (MEF)

E12.5/E14.5Stemcell Technologies Catalog

#00321Catalog#00322

J. Higelin et al. Stem Cell Research 30 (2018) 150–162

153

Page 6: Stem Cell Research, 30: 150-162 Citation for the or iginal ...umu.diva-portal.org/smash/get/diva2:1237422/FULLTEXT01.pdf · This is the published version of a paper published in Stem

Rat embryonic fibroblasts (REF) isolated from day E14 Sprague Dawley rat embryos as described in (Takahashi& Yamanaka, 2006)

N/A

Lenti-X 293T cells Clontech Cat.#632180iPSC line CNTL1 Previously reported in (Stockmann et al., 2013) N/AiPSC line CNTL2 Institute for anatomy and cell biology

Albert-Einstein-Allee 11, 89069 Ulm, GermanyN/A

iPSC line C9ORF Institute for anatomy and cell biology N/AiPSC line NEK1 Institute for anatomy and cell biology N/A

Experimental Models: Organisms/Strains

OligonucleotidesPrimer Hs_HMBS_1_SG Qiagen QuantiTect Primer Assay QT00494130Primer Hs_NEFH_1_SG Qiagen QuantiTect Primer Assay QT00209181Sequencing primer NEK1 fw1 (

CATGAAGATGCCAAAGAGCA)http://www.mwg-biotech.com

Sequencing primer NEK1 fw2 (CTGATACCCGGGAAACTTCA)

http://www.mwg-biotech.com

Recombinant DNApolycistronic vector (for Oct4, Sox2, Klf4, c-

Myc)(Warlich et al., 2011) N/A

pMD2 vector Addgene Cat.#12259psPAX2 vector Addgene Cat.#12260

Software and AlgorithmsAxiovision Rel. 4.8 Zeiss N/AGraph Pad Prism 5 Microsoft N/AFiji (Schindelin et al., 2012) N/ARotor-Gene Q 2.0.2 Qiagen N/AGelAnalyzer http://www.gelanalyzer.com/download.html N/AGIMP 2 https://www.gimp.org/downloads/ N/AAdobe Photoshop CS2 9.0 Microsoft N/AAdobe Illustrator CS2 12.0.0 Microsoft N/AMendeley Desktop 13.1 Mendely Ltd. N/AMicrosoft Office 2013 Microsoft N/A

Other

3. Results and discussion

3.1. Generation of NEK1/C9orf72-mutant hiPSCs and motoneurondifferentiation

To gain a deeper insight into the pathomechanisms triggered byNEK1 loss-of-function mutations in ALS, we analyzed human iPS cellsand differentiated motoneurons (MNs) derived from a patient carryingNEK1c.2434A>T mutation (Brenner et al., 2016).

In this study, we compared newly generated NEK1c.2434A>T (Fig.S2A) with two healthy controls: cell line CNTL1 was previously gen-erated by our group (Higelin et al., 2016), while line CNTL2 was newlycreated from keratinocytes obtained from the daughter of the NEK1-ALS patient. This provided a control cell line, having a genetic back-ground closely related to the mutant cell line. Since the correlation

between NEK1 mutations and ALS has only recently been described, weincluded also a hiPSC line derived from an ALS patient carrying a repeatexpansion of the hexanucleotide (G4C2)1800 within C9orf72 gene (Fig.S2A). This mutation is the most frequent genetic cause of fALS(DeJesus-Hernandez et al., 2011) and previously published studies haveshown that hiPSC derived neurons expressing ALS related C9orf72mutation resemble pathological phenotypes (Lopez-Gonzalez et al.,2016). For these reasons, we included line C9orf72 as a “disease con-trol” for pathologic phenotypes, in order to identify the alterations in-duced by mutation in NEK1 (Fig. 1A).

All newly generated cell lines fulfilled stem cell criteria by expres-sing typical pluripotency markers, having the ability to spontaneouslydifferentiate into the three germ layers (ectoderm, endoderm and me-soderm) (Fig. S1A/B) and presenting normal karyotypes after repro-gramming (Fig. S1C). Neuronal differentiation of iPSCs was achieved as

J. Higelin et al. Stem Cell Research 30 (2018) 150–162

154

Page 7: Stem Cell Research, 30: 150-162 Citation for the or iginal ...umu.diva-portal.org/smash/get/diva2:1237422/FULLTEXT01.pdf · This is the published version of a paper published in Stem

(caption on next page)

J. Higelin et al. Stem Cell Research 30 (2018) 150–162

155

Page 8: Stem Cell Research, 30: 150-162 Citation for the or iginal ...umu.diva-portal.org/smash/get/diva2:1237422/FULLTEXT01.pdf · This is the published version of a paper published in Stem

previously described (Hu and Zhang, 2009). The protocol to inducespecific motoneuronal differentiation includes embryoid bodies (EB)formation and manual selection of neuroepithelial rosettes. After finalplating, 42 days were required to obtain mature MNs (Fig. 1B). In thesecultures about 30% of all cells were chAT+ MNs (Fig. 1C/D). Of note,ALS-related mutations did not significantly affect the differentiationprocedure. Differentiation efficiency was comparable between controlsand mutant lines (no significant differences) and mRNA levels of neu-rofilament heavy chain (NF-H) were similar in all motoneuronal cul-tures (Fig. 1E).

The C9orf72 line maintained the (G4C2)1800 repeat expansion asshown by Southern Blot analysis, revealing the presence of the wildtype gene (green arrow) and the mutated gene (red arrow) in blood(lane1) and in iPSC sample after reprogramming (lane2), although therepeats expansion was slightly shorter in iPSCs compared to the bloodcells (Fig. 1F). Moreover, cells showed characteristic nuclear RNA focihighlighted by fluorescent in situ hybridization (FISH) with a labelledprobe against the hexanucleotide repeat (G4C2) (Fig. 1F). These resultsconfirmed the presence of the pathogenic G4C2 repeat expansion, thusexcluding a possible repeat instability described for C9orf72 (Almeidaet al., 2013). We also confirmed that the mutant NEK1 line conservedthe heterozygous loss of function mutation 2434A > T (p.Arg812Ter)within the NEK1 gene, leading to a premature stop codon as indicatedby red asterisk (Fig. S2A). As expected, the mutation was not detectablein the CNTL2 line, which was derived from the patient's daughter(Fig. 1G, Fig. S1D).

As the heterozygous mutation 2434A > T leads to a premature stopcodon, the mutant cell line was characterized by reduced amount ofNEK1 protein. Western Blot analysis of whole cell lysates from iPSC andMNs showed a reduction up to 50% of NEK1 protein in the mutantgenotype to both controls at hiPSC and MN level (Fig. 1H). Our studyconfirmed previous findings that have indicated NEK1 haploinsuffi-ciency as a pathogenic consequence of NEK1 mutation (Brenner et al.,2016). Since no previous work has described the cellular localization ofNEK1 protein in ALS-related hiPSC, we sought to clarify its compart-mental distribution in this model. In undifferentiated, highly pro-liferative iPSCs, NEK1 localization was mainly restricted to the nucleus,forming protein foci, probably reflecting different proliferation stages(Booth et al., 2014). Interestingly, differentiated, post-mitotic Chat+

MNs were devoid of intranuclear NEK1 but showed a cytoplasmic dis-tribution, suggesting that in MNs NEK1 might also play a role in mi-tochondrial homeostasis control as already observed in immortalizedcells (Chen et al., 2015) (Fig. 1I).

3.2. Increased DNA damage and impaired DNA damage response in ALS-derived motoneurons

Accumulation of DNA breaks and alterations of the DDR machineryhave already been described in familial forms of ALS, more in particularassociated with FUS and C9orf72mutations (Higelin et al., 2016; Lopez-Gonzalez et al., 2016; Qiu et al., 2014; Wang et al., 2013). Since NEK1has been shown to play a key role in DDR (Chen et al., 2008, 2011;Polci et al., 2004), we investigated whether ALS-related NEK1 muta-tions might trigger the accumulation of DNA breaks and alterations inDDR. To this end, we examined the accumulation of γH2A.X+ foci iniPSC (Fig. 2A) and MNs (Fig. 2B) at basal level and after inducing DNAstrand breaks by irradiation. In iPSCs, the basal degree of nuclearγH2A.X foci was comparable amongst all cell lines. The number of cellshaving γH2A.X+ foci was drastically increased after irradiation (1 hafter treatment), and returned to basal level after 24 h. No significantdifference between mutant and control cells was detected, suggestingthat DNA damage can be efficiently repaired by highly-mitotic cellsdespite ALS-related mutations. (Fig. 2C). In contrast, ALS patient-de-rived C9orf72 and NEK1c.2434A>T MNs showed elevated numbers ofγH2A.X+ foci compared to CNTL. The increased number of cells havingγH2A.X+ foci was detectable already at an early stage of neuronaldevelopment (21 days) (Fig. S2B) and this difference became evenlarger at a later stage of maturation (42 days). Moreover,NEK1c.2434A>T cells showed the strongest phenotype when comparedto mutant C9orf72 MNs (C9orf72: 16% ± 1.5, NEK1c.2434A>T:23% ± 1), suggesting an increased cell vulnerability in affected ALSMNs. When challenged with γ-irradiation, all genotypes responded by astrong increase of cells having γH2A.X+ foci after 1 h, withNEK1c.2434A>T MNs displaying the most prominent increase(89% ± 1.6). 24 h after irradiation, mutant and CNTL MNs presentedreduced levels of cells having γH2A.X+ foci compared to the respectiveacute response (1 h after irradiation). However, in contrast to un-differentiated iPSCs, MNs failed to return to basal level, highlightingincreased vulnerability of post-mitotic MNs. Of note, patient-derivedcell lines C9orf72 and NEK1c.2434A>T showed less efficient DDR ma-chinery, as the number of cells displaying γH2A.X+ foci were still in-creased compared to CNTL cells, with the mutant NEK1 cells showingagain the strongest pathological phenotype (Fig. 2D), since C9orf72 atthis stage was only significantly different to CNRL1 but not CNTRL2.

Differences in the response of the two patient cell lines or the twoCNRLs may account for maturation or aging of the cells as previouslyreported (Higelin et al., 2016), and/or human variability and epigenetic

Fig. 1. Characterization of differentiated motoneurons derived from Controls and NEK1/C9orf72 patients.(A) General description of donor cell lines, including gender, age of onset, genotype and clinical information.(B) Time course of the motoneuronal differentiation protocol.(C) Representative immunostainings showing ChAT+ neurons (green) from both controls and both ALS lines.(D) Quantification of ChAT+ neurons in cultures of all generated cell lines. Observed differences between individual cell lines were not statistically significant (one-way ANOVA with Bonferroni post-hoc test, n=3).(E) mRNA expression level of neuronal marker neurofilament H (NF-H). Results are displayed relative to the housekeeping gene HMBS (n= 3).(F) Fluorescent in situ hybridization (FISH) display nuclear RNA foci formation in keratinocytes from C9orf72 patient, not obvious in control. Southern Blot analysisrevealed the presence of either wild type allele (green arrow) or the allele, harboring the typical repeat expansion (red arrow) in blood (lane 1), as well as in hiPSCsample (lane 2).(G) Illustration, showing the DNA and amino acid (AA) sequence of NEK1c.2434A>T and CNTL2, which show defined loss of function mutation c.2434A > T in caseof ALS-patient, leading to a premature stop codon (highlighted by red asterisk).(H) Western Blot analysis of whole cell lysates showed less NEK1 protein in NEK1c.2434A>T compared to both controls in iPSCs and 42 days old MNs. Statisticalanalysis revealed statistical differences between patient and CNTL2 only on iPSC level indicated by asterisk (CNTL2 vs NEK1c.2434A>T ⁎p≤ .05). Statisticallysignificant differences were determined by one-way ANOVA with Bonferroni post-hoc test to compare individual cell lines. Results were normalized to ß-actin anddisplayed relative to untreated CNTL1 (n=3). * p≤ .05, ***p≤ .001, ***p≤ .001.(I) NEK1 immunostainings showing the cellular distribution of the protein (green) in undifferentiated, highly proliferative iPSCs (left) and differentiated, post-mitoticMNs (right), positive for neurofilament H (NF-H). All scale bars: 10 μm.

J. Higelin et al. Stem Cell Research 30 (2018) 150–162

156

Page 9: Stem Cell Research, 30: 150-162 Citation for the or iginal ...umu.diva-portal.org/smash/get/diva2:1237422/FULLTEXT01.pdf · This is the published version of a paper published in Stem

(caption on next page)

J. Higelin et al. Stem Cell Research 30 (2018) 150–162

157

Page 10: Stem Cell Research, 30: 150-162 Citation for the or iginal ...umu.diva-portal.org/smash/get/diva2:1237422/FULLTEXT01.pdf · This is the published version of a paper published in Stem

factors. To analyze whether an aging effect contributes to DNA damageaccumulation, we compared mature 42 days old with young 21 days oldMNs. An age-dependent increase of γH2A.X+ cells was visible in bothcontrols and both patient-derived cell lines, but reached significanceonly in CNTL1 and NEK1c.2434A>T, with the NEK1 mutant showing thestrongest increase (Fig. 2E). However since we detected that inγH2A.X+ cell the number of foci differed, the extent of age-dependentDNA damage accumulation was classified according to the number ofnuclear γH2A.X foci within the nuclei in four categories: (I) no γH2A.Xfoci, (II)< 5 foci, (III) 5–10 foci and (IIII) > 10 foci (Fig. 2F). CategoryIII, presenting a high amount of DNA damage, was slightly increased inpatient-derived cell lines and significant different to CNTL2 duringearly neuronal development. In 42 days old MNs, both patient-derivedcells showed high level of DNA damage (Category III) with significantdifferences to both controls for line NEK1c.2434A>T (Fig. 2G). Thissupports the hypothesis of an affected DDR mechanism in ALS patient-derived MNs, which is in addition, altered during aging. This phenotypeis stronger in the presence of an ALS-related NEK1 mutation suggestingthat NEK1 mutated cells show a more activated DDR at baseline andafter DNA damage stimuli (Chen et al., 2011) than mutated C9orf72.

To finally dissect the extend of DNA breaks accumulation in ALSMNs we performed comet assays (Fig. 3A) and quantified the tail length(Fig. 3B) and the percentage of tail intensity (Fig. 3C) generated byDNA damage. In this assay, control and patient-derived lines showedsimilar results before irradiation but a significant increase of DNA da-mage was seen in NEK1c.2434A>T MNs 24 h after irradiation. Thishighlights the increased vulnerability of the NEK1 mutant cells not onlyin comparison to controls, but also to ALS-related C9orf72 mutants,whose parameters were comparable to the healthy genotype. Con-versely to our results increased levels of DNA damage were previouslyshown in an maturation dependent manner for C9orf72 MNs (Lopez-Gonzalez et al., 2016). For C9orf72 it has already been demonstratedthat the production of dipeptide repeats (DPR) compromises mi-tochondrial function and increases oxidative stress leading to DNAdamage accumulation (Lopez-Gonzalez et al., 2016). This patho-mechanism could not be verified in our analysis when we analyzed thedegree of free radicals in C9orf72 and NEK1 mutants (Fig. S2C).However, this divergence in the analysis of C9orf72 cell lines might beexplained by the differentiation method and the maturation stages. Ouroldest cultures were 6 weeks old as opposed to the study by Lopez and

colleagues, in which significances in DNA damage in mutant c9orf72MNs was observed at 8 weeks in culture. Divergences in NEK1 versusC9orf72 may reflect different pathomechanism and dysregulation ofkey cellular pathways (Gao et al., 2017).

Unrepaired DNA damage leads to chromosomal breaks and to theaccumulation of genomic rearrangements (Khanna and Jackson, 2001)if key components of DDR pathways are affected. Finally, this mightthen accumulate and cause neurodegeneration and neuronal cell death.Our results indicate that mature MNs accumulate DNA damage, how-ever NEK1c.2434A>T fail to repair induced DNA damage leading to anincreased amount of DNA strand breaks visible 24 h after treatment.The fact that this phenotype is detectable by comet assay inNEK1c.2434A>T but not in C9orf72 further suggests that accumulatedand unrepaired DNA damage is more clearly seen in NEK1 mutatedcells, at least at that time point of analysis. In addition, in motoneuronalcultures, caspase-3 levels were also up-regulated only in NEK1c.2434A>T

24 h after irradiation (Fig. 3D). To confirm apoptotic behavior afterDNA damage induction, we quantified the amount of caspase-3 acti-vation, full length protein showed no differences amongst patient andcontrol, however, analysis of active caspase-3 revealed an increase incleaved caspase-3 (17 kDa/19 kDa) 24 h after irradiation inNEK1c.2434A>T, indicating increased vulnerability of patient-derivedcells. As a consequence this finally leads to motoneuronal cell death inneurons carrying ALS-related NEK1 mutation c.2434A > T in responseto DNA damage.

3.3. NEK1 mutated motoneurons are characterized by a misregulation ofDNA damage response machinery

The main DDR pathways that are activated by DNA damage inducedby irradiation are initiated by a complex phosphorylation cascadeleading to recruitment and accumulation of various repair factors atDNA damage foci sites. DNA double-strand break (DSB) repair requireson the one hand the phosphorylation of γH2A.X by ATM serine/threonine kinase (ATM) and Ataxia telangiectasia and Rad3 related(ATR) (Burma et al., 2001). These factors subsequently recruit a largenumber of different mediators, regulating the activation and the loca-lization of BRCA1 to DNA damage foci (Wu et al., 2009). In response,BRCA1 is phosphorylated (pBRCA1) and induces cell cycle checkpointactivation (Fabbro et al., 2004; Xu et al., 2001) and DNA repair (Scully

Fig. 2. Motoneurons derived from ALS-patients show signs of increased DNA damage and impaired DNA damage repair after irradiation and during aging.(A,B) Immunostaining of DNA damage marker γH2A.X (green) in human (A) iPSCs and (B) 42 days old MNs from CNTL1, CNTL2, C9orf72 and NEK1c.2434A>T. Cellswere stained either without irradiation or 1 h/24 h after irradiation. All scale bars: 10 μm.(C) Quantification of γH2A.X+ cells on hiPSC level (n=3). All four lines responded nearly similar to irradiation 1 h and 24 h after irradiation as indicated by redasterisks (CNTL2 Ø vs CNTL2 1 h p≤ .001, C9orf72 Ø vs C9orf72 1 h p≤ .01, NEK1c.2434A>T Ø vs NEK1c.2434A>T 1 h p≤ .001, vs NEK1c.2434A>T 1 h vsNEK1c.2434A>T 24 h p≤ .01).(D) Quantification of γH2A.X+ cells at a later time point of MN maturation (MNs 42 days) without irradiation or 1 h/24 h after irradiation (n=3). All cell linesrespond with a significant increase in γH2A.X+ cells 1 h after irradiation and a subsequent decrease 24 h after treatment as indicated by red asterisks (for all lines: Øvs 1 h p≤ .001, 1 h vs 24 h p≤ .001). Patient-derived C9orf72 and NEK1c.2434A>T presented higher amounts of γH2A.X+ cells under control conditions withouttreatment (CNTL1 vs NEK1c.2434A>T p≤ .001, CNTL2 vs NEK1c.2434A>Tp≤ .05, CNTL1 vs C9orf72 p≤ .05). 1 h after irradiation NEK1c.2434A>T displayed higherdegrees of damaged cells compared to all other lines (CNTL1 vs NEK1c.2434A>T p≤ .001, CNTL2 vs NEK1c.2434A>T p≤ .05, C9orf79 vs NEK1c.2434A>T p≤ .01,CNTL1 vs CNTL2 p≤ .01). After irradiation (24 h) both patients-derived cell lines presented a significant increased γH2A.X+ cell number compared to CNTLs(CNTL1 vs NEK1c.2434A>Tp≤ .001, CNTL2 vs NEK1c.2434A>T p≤ .001, CNTL2 vs C9orf72 p≤ .001, CNTL1 vs CNTL2 p≤ .05). Statistically significant differenceswere determined by two-way ANOVA with Bonferroni post-test.(E) Maturation-dependent increase γH2A.X+ cells in both controls, C9orf72 and NEK1c.2434A>T. Accumulation of DNA damage increased in all lines during ma-turation, but showed only significant differences for CNTL1 and NEK1c.2434A>T (CNTL1 21 days vs 42 days p≤ .05, NEK1c.2434A>T 21 days vs 42 days p≤ .01).Furthermore, the overall level of DNA damage was increased in patient-derived cell lines. Line NEK1c.2434A>T showed the largest response with respect to ma-turation. Unpaired t-test was performed to compare 21 days versus 42 days (n=3).(F) Classification of cells according to the number of nuclear γH2A.X+ foci (grey arrow) as correlation of the degree of DNA damage (red arrow) in the followingcategories: (I) no foci (II) low amount of damage (< 5 γH2A.X foci) (III) 5–10 γH2A.X foci (IIII) high amount of damage (> 10 γH2A.X foci).(G) Distribution of categorized DNA damage depending on the maturation state of neurons. Category III (high amount of damage) was only increased slightly inpatient-derived cell lines in 21 days old MNs compared to CNTL1. 42 days old MNs, derived from patients showed increased amounts of cells, displaying high level ofDNA damage compared to both controls (21 days: CNTL1 vs C9ORF p≤ .001, CNTL2 vs C9ORF p≤ .01, CNTL1 vs NEK1c.2434A>Tp≤ .01, 42 days: CNTL1 vsNEK1c.2434A>Tp≤ .001, CNTL2 vs NEK1c.2434A>Tp≤ .01). Statistically significant differences between distribution patterns were determined by two-way ANOVAwith Bonferroni post-test (n=3). Green stars indicate significance against CNTL1, blue stars indicate significance against CNTL2. * p≤ .05, ***p≤ .001,***p≤ .001.

J. Higelin et al. Stem Cell Research 30 (2018) 150–162

158

Page 11: Stem Cell Research, 30: 150-162 Citation for the or iginal ...umu.diva-portal.org/smash/get/diva2:1237422/FULLTEXT01.pdf · This is the published version of a paper published in Stem

Fig. 3. ALS related NEK1 mutation leads to increased accumulation of DNA breaks in mature motoneurons 24 h after irradiation.(A) Representative images, showing that DNA strand breaks are more abundant in mature MNs of NEK1c.2434A>T 24 h after DNA damage induction as shown bycomet assay.(B, C) Quantification of tail length and percentage of tail intensity from control and patients in 42 days old motoneuronal cultures without and after irradiation. Asignificant increase in accumulated DNA damage was visible 24 h after treatment for NEK1c.2434A>T compared to other cell lines (Tail length: CNTL1 vsNEK1c.2434A>T. p≤ .01, CNTL2 vs NEK1c.2434A>T. p≤ .01, C9orf72 vs NEK1c.2434A>T. p≤ .001, Tail intensity: CNTL1 vs NEK1c.2434A>T. p≤ .001, CNTL2 vsNEK1c.2434A>Tp≤ .01, C9orf72 vs NEK1c.2434A>T. p≤ .01). Statistically significant differences were determined by two-way ANOVA with Bonferroni post-test(n=3). * p≤ .05, ***p≤ .001, ***p≤ .001.(D) Caspase-3 activity of CNTL1, CNTL2, C9orf72 and NEK1c.2434A>T without and 1 h/24 h after irradiation showed that cultures derived from NEK1c.2434A>T

showed higher Caspase-3 activity 24 h after irradiation compared to all other cell lines. This trend showed only significant differences compared to CNTL2 (CNTL2 vsNEK1c.2434A>Tp≤ .05). Statistically significant differences were determined by two-way ANOVA with Bonferroni post-test (n=3). * p≤ .05, ***p≤ .001,***p≤ .001.(E) Western Blot analysis of all four lines 24 h after DNA damage induction showed an increase in caspase-3 cleavage to activated cl.casp-3 (19 kDa) and cl.casp-3(17 kDa) in NEK1c.2434A>T. The amount of full length protein was not affected amongst patient and control. Values are shown relative to β-actin.

J. Higelin et al. Stem Cell Research 30 (2018) 150–162

159

Page 12: Stem Cell Research, 30: 150-162 Citation for the or iginal ...umu.diva-portal.org/smash/get/diva2:1237422/FULLTEXT01.pdf · This is the published version of a paper published in Stem

et al., 1999) via non-homologous end joining (NHEJ) and homologousrecombination (HR). NHEJ, the main radiation-induced DSB repairmechanism, is also initiated through the recruitment of a Ku70/Ku80heterodimer (Fell and Schild-Poulter, 2012). To determine DNA repairmechanism affected in ALS-patient-derived cells, we first quantified theamount of pBRCA1/BRCA1, KU70 and pATR/ATR in mature moto-neuronal cultures before and after induction of DNA damage.

Interestingly, the total amount of cellular BRCA1 was nearly identical inall cells (Fig. 4A), however mutated C9orf72 and NEK1c.2434A>T celllines showed a clear trend towards increased amounts of pBRCA1 underphysiological conditions compared to both controls. After induction ofDNA damage, cells responded with a slight increase in pBRCA1 and thisremained higher in patient-derived lines C9orf72 and NEK1c.2434A>T

24 h after irradiation (Fig. 4A). The amount of DNA damage marker

(caption on next page)

J. Higelin et al. Stem Cell Research 30 (2018) 150–162

160

Page 13: Stem Cell Research, 30: 150-162 Citation for the or iginal ...umu.diva-portal.org/smash/get/diva2:1237422/FULLTEXT01.pdf · This is the published version of a paper published in Stem

KU70 that is implicated in a DNA repair pathway independent ofγH2A.X and BRCA1 remained nearly unaffected in control and patient-derived cells without treatment. After irradiation, KU70 levels slightlydecreased similarly in all cell lines (Fig. 4B). pATR level were almostequal amongst patients and control before and after irradiation(Fig. 4C).

Subsequently, to understand which mechanism or pathways areaffected by mutated NEK1, we employed a specific phospho-proteinarrays to screen for expression changes in cell cycle control and DDRproteins. In NEK1c.2434A>T, we could observe a differential regulation(mainly downregulation) of specific proteins, indicating an impairmentin phosphorylation of some key regulatory elements (Fig. 4D). In ad-dition to increased phosphorylation of BRCA1 and γH2A.X, which areearly downstream events in the ATM-dependent signaling response andwhich require ATM-induced activation (Chen et al., 2011; Gatei et al.,2000), we saw in consensus with the literature an impaired phos-phorylation of cell cycle checkpoint kinase ChK2 (Fig. 4E) inNEK1c.2434A>T. Moreover, we could detect a mild decrease in thephosphorylation of the regulatory proteins p53 (Fig. 4F), ChK1 andsmc1 (Fig. S2D), which are all key elements involved in proper cellcycle control and regulated by ATM (Banin et al., 1998; Edalat, 2012;Yazdi et al., 2002). This is in support of the hypothesis, that when earlyATM-dependent elements are impaired (γH2A.X, pBRCA1) furtherdownstream events of ATM-dependent pathways are also slightlycompromised in NEK1c.2434A>T.

Furthermore, we found phosphorylated myc to be significantly de-creased in NEK1c.2434A>T (Fig. 4G), which is also required for activa-tion of ATM-dependent pathways in response to DNA damage (Guerraet al., 2010), as seen by the phosphorylation of p53 and foci formationof yH2A.X and smc1 (Pusapati et al., 2006). These data suggest thatimpaired myc phosphorylation together with decreased phosphoryla-tion of ChK2/1, p53 and smc1 reflects a complex misregulation of theATM-dependent DDR pathway in the patient line.

In summary, our study provides good evidence that a NEK1 muta-tion associated with ALS leads to NEK1 haploinsufficiency in humanMNs and to increased DNA damage leading to increased vulnerabilityand motoneuronal death. The cellular function of NEK1 is largely un-known, however, its role in early DDR as an essential regulatory kinaseis well documented (Polci et al., 2004) and NEK1−/− cells are char-acterized by unrepaired DNA damage and chromosomal breaks (Chenet al., 2008, 2011). In mutated NEK1c.2434A>T MNs γH2A.X was up-regulated and localized to DNA damage foci and phosphorylation ofBRCA1 was present in MNs after irradiation. This indicates that reduced

levels of NEK1 are still sufficient to activate the DDR pathway. Whenthe system was challenged by an external induction of DNA damage, weclearly saw a less efficient DNA damage repair, leading to accumulationof DNA damage, increased DNA strand breaks and finally to cell death.The response to DNA damage is based on many cellular pathways ac-tivating a large number of different cellular proteins (Shiloh, 2003). Inline with the notion that NEK1 is required for efficient ChK1/ChK2activation (Chen et al., 2008) within ATM pathway, our data show thatNEK1 deficiency triggers a complex misregulation of a complex phos-phorylation pathway. This involves the reduced activation of furtherkey elements, targets of ATM kinase activity (i.e. ChK1/2, p53). Thisresults are in consensus with findings, showing a defective ATM-mediated DNA repair as pathological consequences of ALS associatedC9orf72 expansions (Walker et al., 2017).

Since altered DNA repair mechanisms are potent driving forces forthe progression of neurodegenerative diseases (Higelin et al., 2016;Lopez-Gonzalez et al., 2016; Qiu et al., 2014; Walker et al., 2017; Wanget al., 2013), they should be taken into account when designing newtherapeutic targets to tackle ALS related neurodegeneration.

Supplementary data to this article can be found online at https://doi.org/10.1016/j.scr.2018.06.005.

Acknowledgements

The authors would like to thank Sabine Seltenheim and Lilli Dietzfor excellent technical support. Antje Knehr, Department of Neurology,Ulm University, for collecting human material. Sabine Goll, Departmentof Human Genetics, Ulm University, for karyotyping hiPS cell lines.Benjamin Mayer, Institute for Epidemiology and Medical Biometry, UlmUniversity, for advice in statistical analysis. Pierre-Olivier Frappart,internal medicine, Ulm University, for greatful advice. We are gratefulto all the participants for willingly providing us with hair samples.

Author's contributions

TMB, MD, JH and AC designed and outlined the study. JH and ACcarried out experiments and performed the data analysis. LLSS repro-grammed/characterized cell line C9orf72 and performed FISH experi-ments. SO performed and analyzed irradiation experiments on iPSClevel for CNTL2 and NEK1. AKL contributed to data analysis and sta-tistics. JB and RS performed and analyzed comet assay. GB analyzedkaryotypes. PMA provided keratinocytes. JH, MD and TMB wrote themanuscript.

Fig. 4. Increased phosphorylated BRCA1 in motoneurons derived from ALS-patients.(A-C) Western Blot Analysis of whole cell lysates from mature 42 days old MNs (n=3) showing the protein amount of either (A) phosphorylated Breast Cancer 1(pBRCA1/BRCA1), (B) KU70 and (C) phosphorylated Ataxia telangiectasia and Rad3 related (pATR/ATR) in all four cell lines without treatment, 1 h and 24 h afterirradiation. Results were normalized on β-actin and displayed relative to untreated CNTL1.(A) BRCA1 level was nearly identical amongst all cell lines before and after irradiation.C9orf72 and NEK1c.2434A>T displayed higher levels of pBRCA1 withouttreatment compared to both controls. Protein amount slightly increased 1 h after irradiation in all lines, but remained upregulated in patient cells 24 h after DNAdamage induction, although significant statistical levels were not reached by two-way ANOVA with Bonferroni post-test.(B) Ku70 level was the same amongst all cell lines, but slightly decreased equally after irradiation for CNTL1, CNTL2, C9orf72 and NEK1c.2434A>T.(C) ATR level was nearly identical amongst all cell lines before and 1 h after irradiation, however, C9orf72 showed higher levels 24 h after treatment. Protein amountof phosphorylated ATR (pATR) slightly increased 1 h after irradiation for CNTL1, C9orf72 and NEK1c.2434A>T but showed no differences between patients and CNTLafter DNA damage induction.(D) Heatmap showing phosphorylation levels of proteins involved during cell cycle control and DNA damage response in CNTL and NEK1c.2434A>T in 42 days oldMNs (n=3). Results were displayed relative to control. An overall downregulation of phosphorylated proteins was visible in NEK1 mutated cells. Statisticalsignificant differences and alterations close to statistically significance were obtained by unpaired t-test to compare CNTL and NEK1c.2434A>T as indicated by redarrows.(E) Checkpoint kinase 2 (ChK2) phosphorylated on Ser516 (p= .0548) and Thr68 (p= .0741) was decreased in NEK1c.2434A>T compared to CNTL1. Results wereclose to being statistically significant and determined by unpaired t-test to compare CNTL and mutated cells.(F) p53-protein showed less phosphorylation on Ser15 (p= .1574), Ser33 (p= .1923) and Ser315 (0.1795) in mutated cells compared to CNTL1. Results were closeto being statistically significant and determined by unpaired t-test.(G) Myc-protein phosphorylated on Thr358, Ser373 and Thr58 was decreased in NEK1c.2434A>T compared to CNTL1 (Thr358 p≤ .05, Ser373 p≤ .05, Thr58p= .0658). Statistically significant differences were determined by unpaired t-test (* p≤ .05).(H) Retinoblastoma protein (Rb) showed less phosphorylation on Ser780 (p= .0650), Ser807 (p= .1168) and Ser759 (p= .5248) NEK1c.2434A>T compared toCNTL1. Results were close to being statistically significant and determined by unpaired t-test.

J. Higelin et al. Stem Cell Research 30 (2018) 150–162

161

Page 14: Stem Cell Research, 30: 150-162 Citation for the or iginal ...umu.diva-portal.org/smash/get/diva2:1237422/FULLTEXT01.pdf · This is the published version of a paper published in Stem

Funding

This work was supported by Virtual Helmholtz Institute within theHelmholtz Gesellschaft (“RNA Dysmetabolism in ALS and FTD,” VH-VI-510 to TMB), the Bundesministeriums für Bildung und Forschung(BMBF) (“MND-NET” 01GI0704 to TMB and ACL and 01EK1611C toTMB and MD) and the European Community's Health SeventhFramework Programmed (FP7/2007–2013) under grant agreement no.259867 (to ACL). Sponsors had no influence on the study design or thecollection, analysis and interpretation of data.

Declaration of interest

None.

References

Aasen, T., Raya, A., Barrero, M.J., Garreta, E., Consiglio, A., Gonzalez, F., et al., 2008.Efficient and rapid generation of induced pluripotent stem cells from human kera-tinocytes. Nat. Biotechnol. 26, 1276–1284. doi. https://doi.org/10.1038/nbt.1503.

Almeida, S., Gascon, E., Tran, H., Chou, H.J., Gendron, T.F., Degroot, S., et al., 2013.Modeling key pathological features of frontotemporal dementia with C9ORF72 repeatexpansion in iPSC-derived human neurons. Acta Neuropathol. 126, 385–399. doi.https://doi.org/10.1007/s00401-013-1149-y.

Banin, S., Moyal, L., Shieh, S., Taya, Y., Anderson, C.W., Chessa, L., et al., 1998. Enhancedphosphorylation of p53 by ATM in response to DNA damage. Science 281,1674–1677.

Boillée, S., Vande Velde, C., Cleveland, D.W.W., 2006. ALS: a disease of motor neuronsand their nonneuronal neighbors. Neuron 52, 39–59. doi. https://doi.org/10.1016/j.neuron.2006.09.018.

Booth, D.G., Takagi, M., Sanchez-Pulido, L., Petfalski, E., Vargiu, G., Samejima, K., et al.,2014. Ki-67 is a PP1-interacting protein that organises the mitotic chromosomeperiphery. Elife 3, e01641. doi. https://doi.org/10.7554/eLife.01641.

Brenner, D., Müller, K., Wieland, T., Weydt, P., Böhm, S., Lule, D., et al., 2016. NEK1mutations in familial amyotrophic lateral sclerosis. Brain 139, e28. doi. https://doi.org/10.1093/brain/aww033.

Burma, S., Chen, B.P., Murphy, M., Kurimasa, A., Chen, D.J., 2001. ATM phosphorylateshistone H2AX in response to DNA double-strand breaks. J. Biol. Chem. 276,42,462–42,467. doi. https://doi.org/10.1074/jbc.C100466200.

Chen, Y., Chen, P., Chen, C., Jiang, X., Riley, D.J., 2008. Never-in-mitosis related kinase 1functions in DNA damage response and checkpoint control. 7, 3194–3201.

Chen, Y., Chen, C.F., Riley, D.J., Chen, P.L., 2011. Nek1 kinase functions in DNA damageresponse and checkpoint control through a pathway independent of ATM and ATR.Cell Cycle 10, 655–663. doi. https://doi.org/10.4161/cc.10.4.14814.

Chen, S., Sayana, P., Zhang, X., Le, W., 2013. Genetics of amyotrophic lateral sclerosis: anupdate. Mol. Neurodegener. 8 (1) doi. https://doi.org/10.1186/1750-1326-8-28.

Chen, Y., Craigen, W.J., Riley, D., 2015. Nek1 regulates cell death and mitochondrialmembrane permeability through phosphorylation of VDAC1. 27, 320–331. doi.https://doi.org/10.1002/nbm.3066.Non-invasive.

Cirulli, E.T., Lasseigne, B.N., Petrovski, S., Sapp, P.C., Dion, P.A., Leblond, C.S., et al.,2015. Exome sequencing in amyotrophic lateral sclerosis identifies risk genes andpathways. Science 347, 1436–1441. doi. https://doi.org/10.1126/science.aaa3650.

DeJesus-Hernandez, M., Mackenzie, I.R., Boeve, B.F., Boxer, A.L., Baker, M., Rutherford,N.J., et al., 2011. Expanded GGGGCC hexanucleotide repeat in noncoding region ofC9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 72, 245–256. doi.https://doi.org/10.1016/j.neuron.2011.09.011.

Edalat, F., 2012. Mechanism of p53 stabilization by ATM after DNA damage. 40,1301–1315. doi. https://doi.org/10.1007/s10439-011-0452-9.Engineering.

Fabbro, M., Savage, K., Hobson, K., Deans, A.J., Powell, S.N., McArthur, G.A., et al., 2004.BRCA1-BARD1 complexes are required for P53Ser-15 phosphorylation and a G1/Sarrest following ionizing radiation-induced DNA damage. J. Biol. Chem. 279 (31),251–31,258. doi. https://doi.org/10.1074/jbc.M405372200.

Fell, V.L., Schild-Poulter, C., 2012. Ku regulates signaling to DNA damage responsepathways through the Ku70 von Willebrand A domain. Mol. Cell. Biol. 32, 76–87. doi.https://doi.org/10.1128/MCB.05661-11.

Gao, F.B., Almeida, S., Lopez-Gonzalez, R., 2017. Dysregulation molecular pathways inamyotrophic lateral sclerosis-frontotemporal dementiaspectrum disorder. EMBO J.16, 2931–2950. doi. 10.15252/embj.201797568.

Gatei, M., Scott, S.P., Filippovitch, I., Soronika, N., Lavin, M.F., Weber, B., et al., 2000.Role for ATM in DNA damage-induced phosphorylation of BRCA1. Cancer Res. 60,3299–3304.

Grabrucker, A.M., Schmeisser, M.J., Udvardi, P.T., Arons, M., Schoen, M., Woodling, N.S.,et al., 2011. Amyloid beta protein-induced zinc sequestration leads to synaptic lossvia dysregulation of the ProSAP2/Shank3 scaffold. Mol. Neurodegener. 6, 65. doi.https://doi.org/10.1186/1750-1326-6-65.

Guerra, L., Albihn, A., Tronnersjö, S., Yan, Q., Guidi, R., Stenerlöw, B., et al., 2010. Myc isrequired for activation of the ATM-dependent checkpoints in response to DNA da-mage. PLoS One 5 doi. https://doi.org/10.1371/journal.pone.0008924.

Higelin, J., Demestre, M., Putz, S., Delling, J.P., Jacob, C., Lutz, A.-K., et al., 2016. FUS

mislocalization and vulnerability to DNA damage in ALS patients derived hiPSCs andaging motoneurons. Front. Cell. Neurosci. 10, 1–21. doi. https://doi.org/10.3389/fncel.2016.00290.

Hu, B.-Y., Zhang, S.-C., 2009. Differentiation of spinal motor neurons from pluripotenthuman stem cells. Nat. Protoc. 4, 1295–1304. doi. https://doi.org/10.1038/nprot.2009.127.

Hübers, A., Marroquin, N., Schmoll, B., Vielhaber, S., Just, M., Mayer, B., et al., 2014.Polymerase chain reaction and Southern blot-based analysis of the C9orf72 hex-anucleotide repeat in different motor neuron diseases. Neurobiol. Aging 35,1214.e1–1214.e6. doi. https://doi.org/10.1016/j.neurobiolaging.2013.11.034.

Khanna, K.K., Jackson, S.P., 2001. DNA double-strand breaks: signaling, repair and thecancer connection. Nat. Genet. 27, 247–254.

Kleger, A., Seufferlein, T., Malan, D., Tischendorf, M., Storch, A., Wolheim, A., et al.,2010. Modulation of calcium-activated potassium channels induces cardiogenesis ofpluripotent stem cells and enrichment of pacemaker-like cells. Circulation 122,1823–1836. doi. https://doi.org/10.1161/CIRCULATIONAHA.110.971721.

Lagier-Tourenne, C., Polymenidou, M., Cleveland, D.W., 2010. TDP-43 and FUS/TLS:emerging roles in RNA processing and neurodegeneration. Hum. Mol. Genet. 19,46–64. doi. https://doi.org/10.1093/hmg/ddq137.

Linta, L., Stockmann, M., Kleinhans, K.N., Böckers, A., Storch, A., Zaehres, H., et al., 2012.Rat embryonic fibroblasts improve reprogramming of human keratinocytes into in-duced pluripotent stem cells. Stem Cells Dev. 21, 965–976. doi. https://doi.org/10.1089/scd.2011.0026.

Lopez-Gonzalez, R., Lu, Y., Gendron, T.F., Karydas, A., Tran, H., Yang, D., et al., 2016.Poly(GR) in C9ORF72-related ALS/FTD compromises mitochondrial function andincreases oxidative stress and DNA damage in iPSC-derived motor neurons. Neuron92, 383–391. doi. https://doi.org/10.1016/j.neuron.2016.09.015.

Polci, R., Peng, A., Chen, P.-L., Riley, D.J., Chen, Y., 2004. NIMA-related protein kinase 1is involved early in the ionizing radiation-induced DNA damage response. CancerRes. 64, 8800–8803. doi. https://doi.org/10.1158/0008-5472.CAN-04-2243.

Pusapati, R.V., Rounbehler, R.J., Hong, S., Powers, J.T., Yan, M., Kiguchi, K., et al., 2006.ATM promotes apoptosis and suppresses tumorigenesis in response to Myc. Proc.Natl. Acad. Sci. U. S. A. 103, 1446–1451. doi. https://doi.org/10.1073/pnas.0507367103.

Qiu, H., Lee, S., Shang, Y., Wang, W.Y., Au, K.F., Kamiya, S., et al., 2014. ALS-associatedmutation FUS-R521C causes DNA damage and RNA splicing defects. J. Clin. Invest.124, 981–999. doi. https://doi.org/10.1172/JCI72723.

Schindelin, J., Arganda-Carreras, I., Frise, E., Kaynig, V., Longair, M., Pietzsch, T., et al.,2012. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9,676–682. doi. https://doi.org/10.1038/nmeth.2019.

Scully, R., Ganesan, S., Vlasakova, K., Chen, J., Socolovsky, M., Livingston, D.M., 1999.Genetic analysis of BRCA1 function in a defined tumor cell line. Mol. Cell 4,1093–1099. doi. https://doi.org/10.1016/S1097-2765(00)80238-5.

Shiloh, Y., 2003. 53BP1 and NFBD1/MDC1-Nbs1 function in parallel interacting path-ways activating ataxia-telangiectasia mutated (ATM) in response to DNA damage.Cancer Res. 63, 8586–8591. doi. https://doi.org/10.1038/nrc1011.

Sommer, C.A., Stadtfeld, M., Murphy, G.J., Hochedlinger, K., Kotton, D.N., Mostoslavsky,G., 2016. iPS cell generation using a single lentiviral stem cell cassette. 27, 543–549.doi. https://doi.org/10.1634/stemcells.2008-1075.iPS.

Speit, G., Rothfuss, A., 2012. The comet assay: a sensitive genotoxicity test for the de-tection of DNA damage and repair. Methods Mol. Biol. 920, 79–90. doi. https://doi.org/10.1007/978-1-61,779-998-3_6.

Stockmann, M., Linta, L., Föhr, K.J., Boeckers, A., Ludolph, A.C., Kuh, G.F., et al., 2013.Developmental and functional nature of human iPSC derived motoneurons. Stem CellRev. 9, 475–492. doi. https://doi.org/10.1007/s12015-011-9329-4.

Takahashi, K., Yamanaka, S., 2006. Induction of pluripotent stem cells from mouse em-bryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676. doi.https://doi.org/10.1016/j.cell.2006.07.024.

Taylor, J.P., Brown, R.H., Cleveland, D.W., 2016. Decoding ALS: from genes to me-chanism. Nature 539, 197–206. doi. https://doi.org/10.1038/nature20413.

Vance, C., Rogelj, B., Hortobágyi, T., De Vos, K.J., Nishimura, A.L., Sreedharan, J., et al.,2009. Mutations in FUS, an RNA processing protein, cause familial amyotrophiclateral sclerosis type 6. Science 323, 1208–1211. doi. https://doi.org/10.1126/science.1165942.

Walker, C., Herranz-Martin, S., Karyka, E., Liao, C., Lewis, K., Elsayed, W., et al., 2017.C9orf72 expansion disrupts ATM-mediated chromosomal break repair. Nat. Neurosci.1–14. doi. https://doi.org/10.1038/nn.4604.

Wang, W.-Y., Pan, L., Su, S.C., Quinn, E.J., Sasaki, M., Jimenez, J.C., et al., 2013.Interaction of FUS and HDAC1 regulates DNA damage response and repair in neu-rons. Nat. Neurosci. 16, 1383–1391. doi. https://doi.org/10.1038/nn.3514.

Warlich, E., Kuehle, J., Cantz, T., Brugman, M.H., Maetzig, T., Galla, M., et al., 2011.Lentiviral vector design and imaging approaches to visualize the early stages ofcellular reprogramming. Mol. Ther. 19, 782–789. doi. https://doi.org/10.1038/mt.2010.314.

Wu, J., Huen, M.S.Y., Lu, L.-Y., Ye, L., Dou, Y., Ljungman, M., et al., 2009. Histoneubiquitination associates with BRCA1-dependent DNA damage response. Mol. Cell.Biol. 29, 849–860. doi. https://doi.org/10.1128/MCB.01302-08.

Xu, B., Kim, S.-T., Kastan, M.B., 2001. Involvement of Brca1 in S-phase and G 2-phasecheckpoints after ionizing irradiation. Mol. Cell. Biol. 21, 3445–3450. doi. https://doi.org/10.1128/MCB.21.10.3445.

Yazdi, P.T., Wang, Y., Zhao, S., Patel, N., Lee, E.Y.-H.P., Qin, J., 2002. SMC1 is adownstream effector in the ATM/NBS1 branch of the human S-phase checkpoint.Genes Dev. 16, 571–582. doi. https://doi.org/10.1101/gad.970702.

J. Higelin et al. Stem Cell Research 30 (2018) 150–162

162


Recommended