Abstract
A hexanucleotide repeat expansion on chromosome 9 open reading frame 72 (C9orf72) is associated with familial Amyotrophic lateral sclerosis (ALS) and a subpopulation of patients with sporadic ALS and frontotemporal dementia (FTD). We used inducible pluripotent stem cells from neurotypic and C9orf72+ (C9+) ALS patients to derive neuronal progenitor cells (NPCs). We demonstrated that C9+ and neurotypic NPCs differentiate into neurons. The C9+ neurons, however, spontaneously reexpressed cyclin D1 after 12 weeks, suggesting cell cycle reengagement. Gene profiling revealed significant increases in senescence associated genes in C9+ neurons. Moreover, C9+ neurons expressed high levels of mRNA for CXCL8, a chemokine overexpressed by senescent cells, while media from C9+ neurons contained significant levels of CXCL8, CXCL1, IL13, IP10, CX3CL1 and reactive oxygen species, which are components of the senescence-associated secretory phenotype. Thus, reengagement of cell cycle-associated proteins and a senescence-associated secretory phenotype, could be fundamental components of neuronal dysfunction in ALS and FTD.
Keywords: frontotemporal dementia, amyotrophic lateral sclerosis, cell cycle reentry, senescence, senescence-associated secretory phenotype
1. Introduction
Amyotrophic lateral sclerosis (ALS) is a disease that primarily targets motor neurons resulting in their degeneration and death. Approximately 16,000 people in the United States have ALS and ~5,000 new cases are diagnosed each year (Mehta et al., 2016; Wagner et al., 2015). ALS patients can exhibit cognitive impairment, behavioral issues, and/or dementia, and ALS exhibits significant genetic and functional overlap with the neurodegenerative disorder, Frontotemporal dementia (FTD) (Lattante et al., 2015). FTD is the second most common presenile dementia after Alzheimer’s disease, with manifestations including behavioral disturbances, disinhibition, cognitive decline and/or language dysfunction (Lattante et al., 2015). Ten to fifteen percent of FTD patients have a family history of FTD (Takada, 2015) and up to 40% of FTD patients have concomitant motor neuron disease or motor system dysfunction (Burrell et al., 2011). A hexanucleotide repeat expansion (GGGGCC) on chromosome 9 open reading frame 72 (C9orf72) has been associated with familial ALS and a segment of sporadic ALS and FTD patients (DeJesus-Hernandez et al., 2011). C9orf72 repeat expansion has also been identified as an atypical cause of other neurodegenerative disorders including Parkinson’s, Creutzfeldt-Jakob and Alzheimer’s diseases (Balendra and Isaacs, 2018), likely reflecting the phenotypic heterogeneity complicating clinic-pathological correlation of neurodegenerative diseases and indicating the pleiotropic nature of C9orf72 expansion in neural death.
In neurotypic individuals, there usually are ≤11 hexanucleotide repeats within the C9orf72 gene (Balendra and Isaacs, 2018; Harms et al., 2013; Rutherford et al., 2012; van der Zee et al., 2013). In contrast, in C9+ALS/FTD patients generally have >30 hexanucleotide repeats; however, some C9+ ALS/FTD patients exhibit expansions ranging from hundreds to thousands of repeats. Critically, these hexanucleotide repeats are transcribed into RNA and can be translated into dipeptide repeat proteins (DPRs) (Ash et al., 2013; Balendra and Isaacs, 2018; Donnelly et al., 2013; Gendron et al., 2013; Lagier-Tourenne et al., 2013; Mizielinska et al., 2013; Mori et al., 2013a; Mori et al., 2013b; Zu et al., 2013). Three main competing mechanisms have been posited for the pathogenic effects of C9orf72: a) loss of C9orf72 protein, b) toxic gain of function from C9orf72 repeat RNA or c) toxic gain of function from DPRs (Balendra and Isaacs, 2018). Recently, patient-derived stem cell models have proven useful in recapitulating several key in vivo pathological features of C9orf72 ALS/FTD. These include: a) RNA foci comprising both the sense and antisense direction repeat-laden mRNA (DeJesus-Hernandez et al., 2011); b) the formation of repeat-associated non-AUG translation (RAN) dipeptides (Ash et al., 2013; Mori et al., 2013b; Wen et al., 2014); c) disruptions in nuclear-cytoplasmic transport (both import and export) (Freibaum et al., 2015; Zhang et al., 2015); and d) glutamate toxicity compared to non-ALS iPS neurons (Zhang et al., 2015).
Most studies on C9orf72 ALS/FTD have focused on motor neurons, while only a few have centered on cortical neurons (Guo et al., 2017). Given that ALS patients can present with FTD symptoms (Ling et al., 2013; Ng et al., 2015) and C9orf72 has been found in other neurodegenerative disorders, such as Alzheimer’s disease, we wanted to establish a relevant cell culture model that more accurately reflected the areas of the ALS/FTD brain affected by neurodegenerative processes. We hypothesized that an in vitro three-dimensional culture system derived from cortical neural progenitor cells (NPCs) generated from C9orf72 ALS patients would provide a powerful in vitro model for ALS/FTD, one which more closely mimics native brain morphology and pathology than standard two-dimensional cultures. Therefore, we developed and interrogated such a system, and uncovered spontaneous ectopic cell cycle re-entry and concomitant expression of soluble factors linked with the senescence-associated secretory phenotype in the C9+ iPS neurons, suggesting a novel mechanism for neuronal dysfunction in ALS/FTD.
2. Materials and Methods
2.1. Expansion of inducible pluripotent stem cells (iPSC) and creation of neural progenitor cells.
The following C9+ iPSC lines, CS29iALS-C9n1N (29i) and CS30iALS-C9n1A (30i), were obtained from the Cedars-Sinai Medical Center’s David and Janet Polak Foundation Stem Cell Core Laboratory (Los Angeles, CA) (Supplemental Table 1). Protocols for expanding the C9+ iPSCs were provided by the Cedar-Sinai Medical Center’s David and Janet Polak Foundation Stem Cell Core Laboratory. Briefly, colonies were grown in mTESR™1 medium (Stem Cell Technologies, Cambridge, MA) and passaged until the generation of embryoid bodies (EB). Once at EB stage, all lines were handled as previously described (Brennand et al., 2011) with the exception that neural rosettes and progenitor cells were derived using DMEM/F12+Glutamax medium (ThermoFisher, Waltham, MA) supplemented with 1% N2 (ThermoFisher), 2% B27 without vitamin A (ThermoFisher), 20 μg/mL fibroblast growth factor 2 (FGF2; Peprotech, Rocky Hill, NJ) and 1 μg/mL laminin (Invitrogen, Carlsbad, CA) and were cultured on Matrigel™ (Corning, Corning, NY) coated six-well tissue culture plates. NPCs were stored in liquid nitrogen until use (<12 months). The identity of NPC cultures was confirmed by immunofluorescence (IFC) for Nestin and Sox2, with few cells positive for doublecortin and MAP2.
2.2. NPCs.
The neurotypic control NPC lines, BOH1 and 9319a, were derived from iPSC lines obtained from Dr. K. Brennand (Icahn School of Medicine at Mount Sinai, New York, NY) (Supplemental Table 1). The neurotypic NPC control line, 7545–5b, was derived from cell line GM07545 from the NIGMS Human Genetic Cell Repository at the Coriell Institute for Medical Research (Camden, NJ) and obtained from the University of Virginia Stem Cell Core (Charlottesville, VA) (Supplemental Table 1). All neurotypic NPC control lines were grown and passaged as previously described (Kim et al., 2011).
2.3. NPC differentiation into neurons.
NPCs were thawed and expanded on Matrigel™-coated dishes in NPC medium and split approximately 1:4 every week (up to passage 15) using Accutase (MilliporeSigma, St. Louis, MO). For 2D IFC studies, cells (i.e., passage 10) were seeded at a density of 25 cells/μL onto poly-D-ornithine/laminin-coated coverslips. At 48 hr post-seeding, the culture medium was aspirated and neuron differentiation medium (DMEM/F12+Glutamax, 1% N2, 2% B27 with vitamin A, 20 ng/mL BDNF (Shenandoah Biotechnology, Warwick, PA), 20 ng/mL GDNF (Shenandoah Biotechnology), 500 μM dibutyryl cyclic AMP (MilliporeSigma), 200 nM ascorbic acid (MilliporeSigma), and 1% penicillin-streptomycin (ThermoFisher)) was then added to promote cortical neuronal differentiation. For the first week, differentiation medium was replaced two times. After week one, neuronal differentiation medium supplemented with 1 μg/mL human laminin (Invitrogen) was used as maintenance medium and was replaced at least once per week.
For 3D cultures, Alvetex (ALV) 12-well inserts (ReproCell, Glasgow, UK) were placed into 12-well tissue cell culture plates. Scaffolds were prepared for cell seeding according to manufacturer’s instructions and then coated with 500 μL of human laminin (10 μg/mL) (Invitrogen) for one hr at room temperature (RT). Excess laminin was aspirated from each well. NPCs (i.e., passage 10) were then plated onto each scaffold at a density of 6.28 × 103 cells/μL in 75 μL unsupplemented DMEM/F12+Glutamax medium and allowed to settle into the scaffold for one hr at 37°C. Four mL of NPC medium were then added to each scaffolded well and cultures were incubated for 24 hr. Neuronal differentiation was initiated as described above.
2.4. Cell viability assay.
Calcein AM (CAM) (ThermoFisher) and propidium iodide (PI) (ThermoFisher) were used to assess neuron viability at 8–9 and 43–44 days using both 2D and 3D culturing conditions. For each cell culture condition, three wells were rinsed gently in 37°C Dulbecco’s Phosphate Buffered Saline (DPBS) and then treated with one μM CAM-PI solution in pre-warmed DPBS. Cells were incubated for 30 min at 37°C. CAM-PI was then aspirated and replaced with DPBS at 37°C. Cells were incubated at 37°C for 30 min prior to capturing the images. Three to four images were obtained per culturing condition were taken on a Zeiss 710 Multiphoton Confocal microscope (University of Virginia Advanced Microscopy Core) with a 20x water immersion lens (NA=1.0). Cell counts for the number of live-dead cells were derived using ImageJ (Rueden et al., 2017). A total of 800–1000 cells were counted per replicate per culturing condition.
2.5. Fluorescence immunocytochemistry.
Cells were fixed for 30 min (2D and ALV scaffold) at RT in 4% paraformaldehyde (PFA). Cells were washed three times for 20 min in PBS with 0.1% Tween 20 (PBST). Blocking and permeabilization occurred simultaneously in a solution containing IFC block (2% BSA (Rockland Immunochemicals, Pottstown, PA), 1% fish skin gelatin (MilliporeSigma), 0.02% saponin (Alfa Aesar, Haverhill, MA), 15% horse serum (ThermoFisher) in PBS and 0.3% Triton X-100 (MilliporeSigma) for 4 hr at RT. Primary antibodies (Supplemental Table 2) were diluted in IFC block without Triton X-100, added to permeabilized cells and incubated overnight at 4°C. After incubation, cells were washed twice for 15 min in PBST at RT followed by three washes for 30 min also using PBST at RT. All AlexaFluor (ThermoFisher) secondary antibodies (Supplemental Table 2) were diluted to 2 μg/mL in IF block and incubated for 4 hr at RT. Cells were washed as described above; however, the final wash was with deionized, distilled water (ddH20). Cells were then exposed to DAPI (ThermoFisher, 1:1000 in ddH20) for 10 min at RT, washed three times for 10 min in ddH20 and then mounted on slides with Mowiol (MilliporeSigma). To remove bias towards neuronal markers, we first found scaffolded and 2D fields of view by locating areas of DAPI stained nuclei, and then used other filters to image MAP2, BIII tubulin, tyrosine hydroxylase (TH), and cyclin D1. Images were captured on a Zeiss 710 Multiphoton Confocal microscope as described earlier for the cell viability assay. Cell counts for the number of immunopositive cells are reported as the ratio of immunopositive cells to the total number of DAPI nuclei in an image. Cells counts were derived using ImageJ (Rueden et al., 2017). A total of 150–1500 cells were counted per replicate per culturing condition.
2.6. RNA purification, cDNA synthesis and qPCR.
RNA was purified from each culturing condition as follows. For 2D cell cultures, cells were dissociated with Accutase for 5 min at 37°C, then centrifuged at 1000 × g for 5 min at RT. Cell pellets were washed twice with DPBS. RNA from 2D cell pellets was isolated using an RNeasy kit following the manufacturer’s instructions (Qiagen, Germantown, MD) or flash frozen in liquid nitrogen and stored at −80°C until RNA isolation. For ALV cultures, scaffolds with attached cells were removed from the hanging inserts, placed into a new 12-well plate and washed twice with PBS. Cells were directly lysed with 350 μL RLT lysis buffer with β-mercaptoethanol (1%) for 10 min per manufacturer’s instructions. Cell lysates were passed through a 20-gauge syringe 20 times and then loaded onto a column. The RNeasy protocol was then followed. Real time quantitative PCR was performed with a BioRad CFX 96 using 20 μL duplicate reactions that were made using BioRad SsoAdvanced SYBR green with BIO RAD 20x Primer Assays. mRNA for the housekeeping genes, β-ACTIN, HPRT and/or GAPDH, were used to normalize and calculate ΔΔCTs for gene expression.
Human cell cycle RT2-PCR microarrays were purchased from Qiagen. Total RNA from neurotypic and the C9+ 29i and 30i iPS neurons was purified using an RNeasy Plus RNA isolation kit (Qiagen) per the manufacturer’s instructions. A total of 500–1000 ng of mRNA were converted to cDNA using the RT2 first strand synthesis kit (Qiagen). RT2-PCR microarrays were processed using manufacturer’s instructions. Real-time monitoring of the qPCR reaction was performed on a BioRad CFX Connect thermocycler with RT2 SYBR Green ROX Mastermix (Qiagen). The program was run at 95°C for 10 min and 40 cycles at 95°C for 15 sec and 60°C for one min. RT2-PCR microarray data were analyzed using the Qiagen data webportal (https://www.qiagen.com/us/shop/genes-and-pathways/data-analysis-center-overview-page/). Gene expression was normalized against internal PCR microarray controls (i.e., β-ACTIN, HPRT and GAPDH) using the geometric mean of replicate samples. Significant gene expression was determined by Z-score >1.5. Each cell line under each culture condition was subject to three independent microarray assessments and derived data were used to create volcano plots.
Total RNA was purified from human ALS and ALS/FTD patient brain samples (kind gift from Drs. Timothy Miller and Matthew Harms, Department of Neurology, Washington University in St. Louis, MO) using an RNeasy Plus RNA (Qiagen) isolation kit per the manufacturer’s protocol (Supplemental Table 3). qPCR was performed as described above.
2.7. Luminex analysis of cell culture supernatants derived from iPS neurons cultured on ALV scaffolds.
The quantitative analysis of 41 human chemokines and cytokines was performed by the University of Virginia Flow Cytometry Core Facility. ALV scaffolds were prepared and seeded with neurotypic NPCS (i.e., 7545), 29i or 30i iPS NPCs and differentiated for 12 weeks as described above. At 12 weeks, media from cells were harvested, precleared by centrifugation at 14,000 × g at 4°C, and stored at −80°C until Luminex analysis. Cell supernatants were analyzed for the presence of 41 different cytokines and chemokines using a MILLIPLEX MAP human cytokine/chemokine premixed panel (EMDMillipore, Billerica, MA) using a Luminex MAGPIX system (Luminex, Austin, TX). The analytes were as follows: EGF, eotaxin, FGF-2, Flt-3 ligand, fractalkine, G-CSF, GM-CSF, GRO, IFNα2, IFN-γ, IL-1ra, IL-1α, IL-1β, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12 (p40 and p70), IL-13, IL-15, IL-17A, IP-10, MCP-1, MCP-3, MDC, MIP-1α, MIP-1β, PDGF-AA, PDGF-AB/BB, RANTES, sCD40L, TNF-α, TGF-α, TNF-β, and VEGF.
2.8. Assessment of reactive oxygen species generation.
Cell media supernatants, isolated and prepared from ALV scaffolds as described above, were evaluated for reactive oxygen species (ROS) using ROS-Glo (Promega, Madison, WI) according to manufacturer’s instructions.
2.9. Statistical analyses.
All statistical analyses, including the Benjamini-Hochberg test, were performed with GraphPad Prism 7.0 (GraphPad Software, La Jolla, CA). Data are presented as average (mean) ± SD or SEM. p values were calculated with Student’s t test for comparisons involving two groups or one-way or two-way ANOVA for comparisons involving >two groups. p<0.05 was considered statistically significant. Each experiment is represented by at least three biological replicates and three technical replicates (per independent experiment).
3.0. Reactome analysis.
A Reactome analysis was performed by accessing the https://reactome.org webportal. Common up-regulated and down-regulated genes from the C9+ 29i and 30i versus BOH1 RT2-PCR microarrays were used for the analyses. Pathways most affected by the gene expression patterns were identified and stratified by significance, p-value and false discovery rate (FDR).
3. Results
3.1. C9orf72 (C9+) ALS/FTD iPS cortical neurons express similar neuronal markers as neurotypic controls.
We created iPS NPCs from the patient-derived C9orf72+ iPS cells lines, CS29iALS-C9n1N and CS30iALS-C9n1A, as previously described (Brennand et al., 2011). We fated the NPCs to cortical neurons, the cells most affected in FTD. The derived CS29iALS-C9n1N (29i) and CS30iALS-C9n1A (30i) (i.e., C9+) NPC lines formed neurons using both traditional (i.e., 2D cell culture plastic) and 3D (i.e., ALV scaffolds) cell culture methods. Quantitative PCR analyses at 11 days and 6 weeks post-differentiation showed MAP2 mRNA expression in C9+ 2D and ALV 3D cultures (Figure 1a). Immunofluorescence studies identified MAP2+ neurons in the 29i and 30i 2D and ALV cell models at similar frequencies (Figure 1b, c) and corresponding to levels expressed by neurotypic NPCs by 6 weeks. βIII tubulin and tyrosine hydroxylase expression were also detected at 3 and 6-weeks post-differentiation (Supplemental Figure 1a–b). These data suggested that C9+ iPS neurons displayed similar neuronal differentiation patterns and neuronal markers as neurotypic iPS neurons.
Figure 1. C9+ iPS neurons express neuronal markers in 2D and 3D culture.
Panel a, Map2 gene expression levels by qPCR in neurotypic (i.e., BOH1 and 9319a) and C9+ (i.e., 29i and 30i) iPS neurons. Black bars, 11 days post-differentiation induction. White bars, 6 weeks post-differentiation induction. (--- undifferentiated control). N=2 per cell line. Panel b, MAP2 percent positive cells as detected by immunofluorescence at 6 weeks post-differentiation induction. Panel c, Neurotypic (BOH1) and C9+ iPS neurons after differentiation on ALV scaffolding for 6 weeks. Neurons (MAP2, green), astrocytes (GFAP, pink) and nuclei (DAPI, blue), (20x objective, NA 0.8, scale bar 100μm). Panel d, Viability of C9+ ALS/FTD and neurotypic neurons at 8 days (black bars) and 6 weeks (white bars) post-differentiation induction using calcein AM. N=12 per cell line.
In 2D, neurotypic and C9+ neuronal cultures displayed similar levels of viability at 8 days and 6 weeks (Figure 1d). Noteworthy, however, was the statistically significant decrease in the viability of the 3D C9+ 30i iPS neuronal co-cultures at 8 days and 6 weeks differentiation (Figure 1d). This viability decrease was not observed in the C9+ 29i cultures (Figure 1d). However, with extended culturing in 2D and 3D (>200 days), both C9+ cell lines trended with higher mortality versus neurotypic cells (Supplemental Figure 1d). These data suggested that, although the C9+ iPS neuronal cultures have similar differentiation patterns, they may be characterized by a general loss of fitness versus neurotypic controls. However, we note that the calcein AM viability assessment provides insight to the health of the overall neuronal co-culture and precise cell populations affected are not discernible.
3.2. C9+ iPS neurons spontaneously express cyclin D1 after 12 weeks in 3D culture.
Control neurotypic and C9+ NPCs were seeded in 2D as well as in ALV scaffolds and then differentiated into neurons for up to 3 months. 2D cultures and ALV scaffolds with differentiating neurotypic or C9+ NPCs were then immunostained for expression of cyclin D1, a marker of G1 of the cell cycle, at 6 and 12 weeks after induction of differentiation (Figure 2). Immunostaining of the ALV scaffolds for cyclin D1 protein levels revealed a statistically significant increase in the C9+ versus neurotypic iPS neuronal cultures at 12 weeks post-differentiation (Figure 2b). We conclude that C9+ neurons grown in 3D culture spontaneously re-enter the cell cycle, which interestingly precedes most neuron death in Alzheimer’s disease. In contrast, no such statistically significant difference in cyclin D1 expression for C9+ versus neurotypic iPS neurons was observed in 2D cultures at any time point (Figure 2c); however, the data suggested a similar trend in cyclin D1 expression upon extended culturing.
Figure 2. ALS/FTD C9+ cell lines spontaneously reenter the cell cycle.
Panel a. ALS/FTD C9+ (29i and 30i) and neurotypic cell lines spontaneously express cyclin D1 protein in 3D culture 12-weeks post-differentiation. Shown are representative images of the 9319a and 29i+ cell lines. ALV cultures were stained for IF with anti-MAP2 (green) and anti-cyclin D1 (CYD1, red), which identifies mature neurons and cells that have exited G0 and entered into G1, respectively. Nuclei were stained with DAPI (blue) (20x objective, NA 0.8, scale bar 100μm). Panel b. Quantification of cyclin D1 expression in neurotypic (black bars) and C9+ (white bars) cells grown in 2D and on ALV scaffolding. N=3 (9319a, 29i,) and N=2 (BOH1, 30i) at 6 weeks a minimum of 800 cells was counted total across each replicate; N=2 (9319a, BOH1, 29i and 30i) at 12 weeks; a minimum of 150 cells was counted across each replicate.
3.3. C9+-derived iPS neuronal cultures have dysregulated cell cycle-associated gene expression.
To characterize more fully the observed spontaneous re-expression of cyclin D1 by C9+ iPS neurons at 12 weeks in 3D culture, we harvested RNA from 29i and 30i C9+ and neurotypic (BOH1) iPS neuron cultures at 3-months post-differentiation and performed a cell cycle gene-targeted RT2-PCR array, which comprised 84 genes associated with the mammalian cell cycle. In the 29i C9+ iPS neurons, the expression levels of four genes were upregulated (fold change>1.5) under basal 3D culturing conditions: CDKN2A, CDKN2B, CDKN1A and CCND1 and three genes were downregulated in expression: AURKB, GTSE1 and CCNA2 (Figure 3a). In the 30i C9+ iPS neurons, in contrast, four genes were upregulated (i.e., CDKN2A, CDKN2B, CCNH, and CCND3), while ten genes were downregulated (i.e., AURKB, CDK2, MCM3, KPNA2, MKI67, ATM, CCNB1, CDC20, GTSE1, and CDK1) (fold change >1.5) (Figure 3b).
Figure 3. Volcano plot of relative differences in mRNA gene expression of two C9+ ALS versus neurotypic cells cultured for three months on ALV scaffolding.
Genes significantly overexpressed (fold change >1.5 or <−1.5) are in red (•) with remaining genes being denoted in green (•). The dashed line = p<0.05. N=3 independent microarrays per cell line. BOH1 cell line used as the neurotypic control.
We next confirmed the gene expression of the overlapping gene set (i.e., CDKN2A, CDKN2B, AURKB and GTSE1) (Figure 4) using RNA isolated from 29i C9+ iPS neurons cultured for 3 months on ALV. Benjamini-Hochberg analysis predicted that CDKN2A, CDKN2B and AURKB were true positives while GTSE1 might be a false positive (Supplemental Table 4). Using qRT-PCR, we confirmed CDKN2A and CDKN2B gene expression was upregulated ~20- and ~5-fold, respectively, versus neurotypic control (Figure 4a–b). AURKB downregulation was confirmed in both 29i iPS neuron RNA samples (Figure 4c), while downregulation of GTSE1 gene expression was confirmed in only one of the two 29i iPS neuron RNA samples (Figure 4d) supporting the computational Benjamini-Hochberg analysis (Supplemental Table 4) which suggested it may be a false positive.
Figure 4. qPCR confirmation of CDKN2A, CDKN2B, GTSE1 and AURKB gene expression in C9+ 29i ALS cell line versus BOH1 neurotypic control cell line.
mRNA gene expression levels were normalized relative to ACTIN, HPRT and GAPDH. N=2 biological replicates. N=3 technical replicates per control gene ± SEM. Panel a, CDKN2A. Panel b, CDKN2B. Panel c, GTSE1. Panel d, AURKB. A, replicate 1. B, replicate 2.
3.4. Computational analysis of overlapping gene sets suggest cellular senescence pathways are activated in 29i and 30i C9+ neurons.
To identify possible cellular pathways most affected by the CDKN2A, CDKN2B, and AURKB gene signature in the C9+ iPS neurons, we performed computational analysis using Reactome (Fabregat et al., 2016), which suggested that the cellular responses to stress pathway node was significantly over-represented in the C9+ iPS neurons based on the CDKN2A, CDKN2B and AURKB gene signature (Figure 5a, Supplemental Figure 1). Moreover, there was a specific emphasis on pathways leading to overall cellular senescence, including oncogene induced senescence, oxidative stress induced senescence, cellular senescence and the senescence-associated secretory phenotype (Figure 5a, Table 1).
Figure 5. Reactome analysis of overlapping gene hits from 29i and 30i ALS/FTD C9+ cell lines.
Panel a, Reactome (https://reactome.org) computational analysis of the common gene hits from the qRT-PCR microarrays highlight the potential involvement of cellular senescence pathways in the CCR of the 29i and 30i cell lines after 12 weeks in 3D culture. Panel b, qPCR was performed on RNA from 29i, 30i and 7545 neurotypic control cells to quantify CXCL8 gene expression. Panel c, Luminex analysis was performed for detection of secreted CXCL8. N=3 replicates ± SEM. Error bars=SEM.
Table 1.
Reactome analyses
PATHWAY NAME | p-value | FDR |
---|---|---|
Oncogene induced senescence | 8.33E-7 | 1.52E-4 |
Oxidative stress induced senescence | 2.57E-5 | 2.34E-3 |
Cellular senescence | 1.14E-4 | 6.95E-3 |
Senescence-associated secretory phenotype | 2.47E-3 | 1.11E-1 |
Cellular response to stress | 5.34E-3 | 1.92E-1 |
3.5. ALD/FTD C9+ iPS neurons express components of the senescence-associated secretory phenotype.
To confirm that the 29i and 30i C9+ ALS/FTD iPS neuronal cultures were indeed undergoing senescence at 12-weeks in 3D culture, we performed qPCR on 29i and 30i iPS neuron-derived RNA for detection of IL-6 and/or CXCL8 (IL-8), which are two soluble factors closely associated with cellular senescence (Tan et al., 2014). CXCL8 gene expression was significantly upregulated in 29i and 30i C9+ iPS neurons (Figure 5b) compared to neurotypic controls, while there was no detectable change in IL-6 gene expression (data not shown). Nonetheless, the detection of upregulated CXCL8 gene expression suggested that the C9+ iPS neurons were indeed undergoing senescence. Subsequently, we initiated additional 29i, 30i and neurotypic iPS neuronal cultures in 3D, maintained them in culture for 12 weeks, and then harvested the cell culture media for quantification of secreted cytokines and chemokines by bead-based multianalyte profiling. We confirmed the significant overexpression of components of the senescence-associated secretory phenotype including CXCL8 (IL-8), CCL2 (MCP1), CXCL1 (GROα), IL-13 and IP10 (Figure 5c and Figure 6) in both the 29i and 30i ALS/FTD cell lines versus neurotypic controls. Additionally, CX3CL1 (fractalkine) was also significantly overexpressed in the C9+ ALS/FTD iPS neurons (Figure 6c). Fractalkine has been described as a “find me” (Sokolowski et al., 2014) signal for neurons undergoing ethanol-induced or glutamate-induced (i.e., excito-neurotoxicity) apoptosis as a means to promote efferocytosis through the recruitment of microglia (Noda et al., 2011; Sokolowski et al., 2014). We also detected in the media from 29i and 30i iPS neurons significantly elevated levels of ROS, which are considered inducers of cellular senescence (Tchkonia et al., 2013) (Figure 6f).
Figure 6. Detection of components of the senescence-associated secretory phenotype in cell culture supernatants from ALS/FTD C9+ cell lines.
29i, 30i and 7545 neurotypic control cells were seeded into ALV scaffolds and cultured for 12 weeks. Cell culture supernatants were harvested and assessed by Luminex analysis or for ROS detection. Panel a, CCL2 (MCP1). Panel b, CXCL1 (GROα). Panel c, CX3CL1 (fractalkine). Panel d, IL-13. Panel e, IP-10. Panel f, reactive oxygen species. Panel g, qPCR confirmation of CDKN2A and CDKN2B gene expression changes in human patient samples. ●, C9+. ●, C9−. mRNA levels were normalized relative to GAPDH. N=3 replicates ± SEM. Error bars=SEM. C=normal human brain RNA.
3.6. ALS/FTD patients have upregulated CDKN2A gene expression.
We next examined the gene expression levels of CDKN2A and CDKN2B in ALS and ALS/FTD patient brain RNA samples using qRT-PCR. CDKN2A gene expression was increased on average more than 4-fold in brain samples of patient with ALS and ALS/FTD (Figure 6g), while CDKN2B was unaltered. The range observed in CDKN2A expression levels may be in part dependent upon the time of post-mortem sampling (Supplemental Table 3). However, it would be more expected for RNA degradation to be observed, with extended post-mortem sampling periods, versus increased gene expression.
4. Discussion
The need for more pathologically relevant neurodegeneration models has become obvious with the growing realization of the limitations of mouse and traditional 2D cell culture systems (Centeno et al., 2018; Dawson et al., 2018; Jucker, 2010). Human iPSCs offer more physiologically relevant cell-based populations for experimental studies and can be differentiated into many cell lineages, such as cortical and motor neurons. Critically, patient-derived stem cell models have proven useful in demonstrating several key pathological features of C9orf72 ALS/FTD including: 1) RNA foci comprising both the sense and antisense direction repeat-laden mRNA (DeJesus-Hernandez et al., 2011); 2) formation of repeat-associated non-AUG translation (RAN) dipeptides (Ash et al., 2013; Mori et al., 2013b; Wen et al., 2014); 3) disruption of both import and export for nuclear-cytoplasmic transport (Freibaum et al., 2015; Zhang et al., 2015); and 4) glutamate toxicity compared to non-ALS iPS neurons (Donnelly et al., 2013). In general, however, the impact of iPS cells is most readily observable when cultured in 3D, which allows for the interactions with other cell populations. To that end, several groups have developed 3D iPS neuronal culture platforms (Hopkins et al., 2015; O’Connor et al., 2001; Yoo et al., 2011). Most notably, Choi et al. (Choi et al., 2014), cultured ReN cells in 3D and successfully replicated the Aβ and tau pathologies, as well as the inflammatory microenvironment (Park et al., 2018) found in Alzheimer’s disease, which is not discernible in 2D culture. Here, we demonstrate that differentiating C9+ iPS cells into cortical neurons in 3D is feasible and may help to further phenotyping of C9+ ALS/FTD. Furthermore, our 3D ALS/FTD C9+ iPS neuron-based model system recapitulates the reactivation of cell cycle regulators (i.e., cyclin D1) frequently observed in both neurodegeneration patients and corresponding in vitro models (Di Giovanni et al., 2005; Khan et al., 2018; Kodis et al., 2018; Lim and Qi, 2003; Maccioni et al., 2001; Norambuena et al., 2017; Pelegri et al., 2008; Ranganathan and Bowser, 2003, 2010; Rashidian et al., 2005; Rashidian et al., 2007; Seward et al., 2013).
Unfortunately, in neurodegenerative diseases, the neuronal loss of fitness and death occurs over the span of many years making the specific mode of neuron “loss” (e.g., apoptosis, necrosis, excitotoxicity, excess autophagy, synaptic loss, long-term potentiation impairment, neurite dysfunction, senescence), and any of its originating drivers, challenging to delineate. However, re-expression of cell cycle regulators (e.g., cyclin D1, CDK4, hyperphosphorylated pRb and E2F1) may be a common conduit leading to in vitro and in vivo neuronal dysfunction and death prior to and during neurodegenerative processes. There is significant evidence that neurons in vitro undergo cell cycle reentry in response to a variety of stressors, including neurotrophic factor deprivation (brain-derived neurotrophic factor, glial cell line-derived neurotrophic factor), DNA damage, and oxidative stress (reviewed in (Zhu et al., 2004)). Neurons exposed to stressors usually die at the G1/S checkpoint prior to DNA synthesis; however, in some instances DNA synthesis as visualized by bromodeoxyuridine incorporation has been observed (Seward et al., 2013; Varvel et al., 2008). Recent evidence, however, suggests primary neurons can enter M-phase (albeit with an engineered neuronal cell system) with a small number of neurons actually dividing (Walton et al., 2019), although it is likely neurons more commonly exist in a prolonged 4N state (Frade and Ovejero-Benito, 2015). Nonetheless, these data suggest that neurons retain the cell cycle machinery needed for cell cycle progression (Walton et al., 2019), and with the appropriate stimuli, neurons have the capacity to re-engage the cell cycle.
Neurons may be particularly sensitive to oxidative stress. The “two-hit” model of neurodegeneration (Zhu et al., 2004) suggests that oxidative stress in conjunction with re-engagement of the cell cycle machinery in “at risk” neurons may be necessary for neurodegenerative progression. In this model, cell cycle re-entry and elevated oxidative stress can exist separately (i.e., “single hit”), but when experienced concurrently (“second hit”), the neurodegenerative process initiates (Zhu et al., 2004). Moreover, this model proposes the existence of an “oxidative steady-state” in which “at risk” neurons are chronically bombarded with ROS yet remain functional for years. These neurons are thought to be exquisitely sensitive to the “second hit”, which provides the impetus toward neurodegeneration (LeBel and Bondy, 1992; Zhu et al., 2004). Oxidative stress also triggers cellular senescence, which is defined as irreversible or sustained cell cycle arrest; however, senescence is a complex cellular response to a variety of stressors, including mitochondrial deterioration, ROS, and oncogene expression (Campisi, 2013). Senescence also promotes chronic inflammation, a state referred as “inflammaging” (Franceschi et al., 2018; Kritsilis et al., 2018), which is caused by increased secretion of pro-inflammatory cytokines and other soluble factors. These proinflammatory cytokines often engage signaling effectors associated with the cell cycle or that serve to regulate the expression of cell cycle proteins and may deliver the “second hit” required for neurodegenerative progression (Cingoz and Goff, 2018; Leslie et al., 2006; Mori et al., 2013a; Tian et al., 2017). Notably, we detected multiple proinflammatory chemokines (i.e., CXCL8, CCL2, CXCL1, and IP-10) in our 3D ALS/FTD neuronal cultures as part of the senescence-associated secretory phenotype. Moreover, IL-13, which was also detected in our ALS/FTD neuronal cultures, can sensitize cells to oxidative stress during neuro-inflammatory conditions (reviewed in (Mori et al., 2016)).
Unfortunately, universally accepted biochemical markers of senescence are not yet available, but examples of senescence markers that have been used include p16INK4a and p21 expression, β-galactosidase activity, telomere-associated foci, epigenomic alterations, ROS, DNA damage and the senescence–associated secretory phenotype (SASP) (Myrianthopoulos et al., 2019; Sun et al., 2018). Some frequently used endpoints, such as β-galactosidase activity, have not been useful with neurons (Piechota et al., 2016). Although most commonly associated with cancer and aging, cellular senescence may contribute to, and perhaps drive, neurodegenerative diseases. Several recent studies have demonstrated that senescent cells (i.e., astrocytes, microglia, oligodendrocytes, oligodendrocyte progenitor cells, neurons, and neural stem cells) (reviewed in (Kritsilis et al., 2018)) exist within the brains of aging organisms, as well as in brains with neurodegenerative disease, suggesting that they promote neuronal dysfunction (Musi et al., 2018; Oost et al., 2018) and the associated cognitive decline (Bussian et al., 2018).
If either senescence or cell cycle re-entry are drivers of ALS/FTD pathology, then it is conceivable that compounds that target these processes, such as the senolytic (Myrianthopoulos et al., 2019; Xu et al., 2018) or cell cycle inhibitors (Xu et al., 2013), might have some prophylactic or therapeutic potential for the disease. The 3D iPSC model, which we have developed, might be a valuable tool to help facilitate these studies especially since it comprises neuronal as well as glial cell populations. Thus, in vitro and in vivo models that recapitulate these cell cycle re-entry SASP phenotypes may be invaluable resources to understand the timing and progression of ALS/FTD associated neurodegenerative events.
Supplementary Material
Supplemental Figure 1. Panels a and b, Expression of βIII tubulin and tyrosine hydroxylase (TH) by neurotypic 9319a and C9+ 30i iPS neurons. Panel c, Alexa fluor secondary antibody only control. No primary antibodies were added. Anti-chicken IgY 488, anti-rabbit IgG 568, and anti-mouse IgG 647. Scale bar = 100 μm. Panel d, Viability assessment of neurotypic BOH1, C9+ 29i and C9+ 30i iPS neuronal cultures at 200 days post-differentiation.
Supplemental Figure 2. Venn diagram of differentially expressed cell cycle-associated genes in 29i and 30i iPS neurons.
Supplemental Table 1. C9+ cell lines used for neuronal progenitor cell derivation.
Supplemental Table 2. Antibodies used in studies.
Supplemental Table 3. Patient information.
Supplemental Table 4. Benjamini-Hochberg test on C9+ 29i and 30i p-values. Red text indicates hits from C9+ 29i PCR microarray studies. Bolded red text indicates genes that confirmed in C9+ 29i confirmation studies. Bolded black text indicates additional hits from C9+ 30i PCR microarray studies.
Highlights.
Neurotypic and C9+ NPCs can be differentiated in two- and three-dimensional models.
C9+ NPCs spontaneously expressed cyclin D1 protein in three-dimensional culture.
Expression profiling showed increases in genes associated with cellular senescence.
C9+ neurons expressed factors of the senescence-associated secretory phenotype.
5. Acknowledgments
The work was supported by grants from the Fiske Drug Discovery Fund (J.S.L., E.R.S.), the Owens Foundation (E.F., J.S.L., G.S.B.), NIH RF1 AG51085 (G.S.B.), NIH R01 AG063400 (E.R.S.), Hartwell Foundation (E.F.) and the Cure Alzheimer’s Fund (G.S.B., J.S.L, E.R.S.). This work used the Zeiss 710 Confocal Microscope in the Advanced Microscopy Facility, the Luminex MagPix in the Flow Cytometry Core and neural progenitor cells produced by the Stem Cell Core Facility, which are all facilities supported by the University of Virginia School of Medicine. We also thank the Cedars-Sinai Medical Center’s David and Janet Polak Foundation Stem Cell Core Laboratory for the ALS C9+ iPS cells. This manuscript is dedicated to the memory of James F. Capps (1958-2015).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Disclosure statement
The authors have nothing to disclose. Dr. E. Foff now works for Acadia Pharmaceuticals. Dr. Shahzad Khan now works at Stanford University.
7. References
- Ash PE, Bieniek KF, Gendron TF, Caulfield T, Lin WL, Dejesus-Hernandez M, van Blitterswijk MM, Jansen-West K, Paul JW 3rd, Rademakers R, Boylan KB, Dickson DW, Petrucelli L, 2013. Unconventional translation of C9ORF72 GGGGCC expansion generates insoluble polypeptides specific to c9FTD/ALS. Neuron 77(4), 639–646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Balendra R, Isaacs AM, 2018. C9orf72-mediated ALS and FTD: multiple pathways to disease. Nat Rev Neurol 14(9), 544–558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brennand KJ, Simone A, Jou J, Gelboin-Burkhart C, Tran N, Sangar S, Li Y, Mu Y, Chen G, Yu D, McCarthy S, Sebat J, Gage FH, 2011. Modelling schizophrenia using human induced pluripotent stem cells. Nature 473(7346), 221–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burrell JR, Kiernan MC, Vucic S, Hodges JR, 2011. Motor neuron dysfunction in frontotemporal dementia. Brain 134(Pt 9), 2582–2594. [DOI] [PubMed] [Google Scholar]
- Bussian TJ, Aziz A, Meyer CF, Swenson BL, van Deursen JM, Baker DJ, 2018. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature 562(7728), 578–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Campisi J, 2013. Aging, cellular senescence, and cancer. Annu Rev Physiol 75, 685–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Centeno EGZ, Cimarosti H, Bithell A, 2018. 2D versus 3D human induced pluripotent stem cell-derived cultures for neurodegenerative disease modelling. Mol Neurodegener 13(1), 27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choi SH, Kim YH, Hebisch M, Sliwinski C, Lee S, D’Avanzo C, Chen H, Hooli B, Asselin C, Muffat J, Klee JB, Zhang C, Wainger BJ, Peitz M, Kovacs DM, Woolf CJ, Wagner SL, Tanzi RE, Kim DY, 2014. A three-dimensional human neural cell culture model of Alzheimer’s disease. Nature 515(7526), 274–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cingoz O, Goff SP, 2018. Cyclin-dependent kinase activity is required for type I interferon production. Proc Natl Acad Sci U S A 115(13), E2950–E2959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dawson TM, Golde TE, Lagier-Tourenne C, 2018. Animal models of neurodegenerative diseases. Nat Neurosci 21(10), 1370–1379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M, Rutherford NJ, Nicholson AM, Finch NA, Flynn H, Adamson J, Kouri N, Wojtas A, Sengdy P, Hsiung GY, Karydas A, Seeley WW, Josephs KA, Coppola G, Geschwind DH, Wszolek ZK, Feldman H, Knopman DS, Petersen RC, Miller BL, Dickson DW, Boylan KB, Graff-Radford NR, Rademakers R, 2011. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 72(2), 245–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Giovanni S, Movsesyan V, Ahmed F, Cernak I, Schinelli S, Stoica B, Faden AI, 2005. Cell cycle inhibition provides neuroprotection and reduces glial proliferation and scar formation after traumatic brain injury. Proc Natl Acad Sci U S A 102(23), 8333–8338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Donnelly CJ, Zhang PW, Pham JT, Haeusler AR, Mistry NA, Vidensky S, Daley EL, Poth EM, Hoover B, Fines DM, Maragakis N, Tienari PJ, Petrucelli L, Traynor BJ, Wang J, Rigo F, Bennett CF, Blackshaw S, Sattler R, Rothstein JD, 2013. RNA toxicity from the ALS/FTD C9ORF72 expansion is mitigated by antisense intervention. Neuron 80(2), 415–428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fabregat A, Sidiropoulos K, Garapati P, Gillespie M, Hausmann K, Haw R, Jassal B, Jupe S, Korninger F, McKay S, Matthews L, May B, Milacic M, Rothfels K, Shamovsky V, Webber M, Weiser J, Williams M, Wu G, Stein L, Hermjakob H, D’Eustachio P, 2016. The Reactome pathway Knowledgebase. Nucleic Acids Res 44(D1), D481–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frade JM, Ovejero-Benito MC, 2015. Neuronal cell cycle: the neuron itself and its circumstances. Cell Cycle 14(5), 712–720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Franceschi C, Garagnani P, Parini P, Giuliani C, Santoro A, 2018. Inflammaging: a new immune-metabolic viewpoint for age-related diseases. Nat Rev Endocrinol 14(10), 576–590. [DOI] [PubMed] [Google Scholar]
- Freibaum BD, Lu Y, Lopez-Gonzalez R, Kim NC, Almeida S, Lee KH, Badders N, Valentine M, Miller BL, Wong PC, Petrucelli L, Kim HJ, Gao FB, Taylor JP, 2015. GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport. Nature 525(7567), 129–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gendron TF, Bieniek KF, Zhang YJ, Jansen-West K, Ash PE, Caulfield T, Daughrity L, Dunmore JH, Castanedes-Casey M, Chew J, Cosio DM, van Blitterswijk M, Lee WC, Rademakers R, Boylan KB, Dickson DW, Petrucelli L, 2013. Antisense transcripts of the expanded C9ORF72 hexanucleotide repeat form nuclear RNA foci and undergo repeat-associated non-ATG translation in c9FTD/ALS. Acta Neuropathol 126(6), 829–844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo W, Fumagalli L, Prior R, Van Den Bosch L, 2017. Current Advances and Limitations in Modeling ALS/FTD in a Dish Using Induced Pluripotent Stem Cells. Front Neurosci 11, 671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harms MB, Cady J, Zaidman C, Cooper P, Bali T, Allred P, Cruchaga C, Baughn M, Libby RT, Pestronk A, Goate A, Ravits J, Baloh RH, 2013. Lack of C9ORF72 coding mutations supports a gain of function for repeat expansions in amyotrophic lateral sclerosis. Neurobiol Aging 34(9), 2234 e2213–2239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hopkins AM, DeSimone E, Chwalek K, Kaplan DL, 2015. 3D in vitro modeling of the central nervous system. Prog Neurobiol 125, 1–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jucker M, 2010. The benefits and limitations of animal models for translational research in neurodegenerative diseases. Nat Med 16(11), 1210–1214. [DOI] [PubMed] [Google Scholar]
- Khan SS, LaCroix M, Boyle G, Sherman MA, Brown JL, Amar F, Aldaco J, Lee MK, Bloom GS, Lesne SE, 2018. Bidirectional modulation of Alzheimer phenotype by alpha-synuclein in mice and primary neurons. Acta Neuropathol 136(4), 589–605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim JE, O’Sullivan ML, Sanchez CA, Hwang M, Israel MA, Brennand K, Deerinck TJ, Goldstein LS, Gage FH, Ellisman MH, Ghosh A, 2011. Investigating synapse formation and function using human pluripotent stem cell-derived neurons. Proc Natl Acad Sci U S A 108(7), 3005–3010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kodis EJ, Choi S, Swanson E, Ferreira G, Bloom GS, 2018. N-methyl-D-aspartate receptor-mediated calcium influx connects amyloid-beta oligomers to ectopic neuronal cell cycle reentry in Alzheimer’s disease. Alzheimers Dement 14(10), 1302–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kritsilis M, S VR, Koutsoudaki PN, Evangelou K, Gorgoulis VG, Papadopoulos D, 2018. Ageing, Cellular Senescence and Neurodegenerative Disease. Int J Mol Sci 19(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lagier-Tourenne C, Baughn M, Rigo F, Sun S, Liu P, Li HR, Jiang J, Watt AT, Chun S, Katz M, Qiu J, Sun Y, Ling SC, Zhu Q, Polymenidou M, Drenner K, Artates JW, McAlonis-Downes M, Markmiller S, Hutt KR, Pizzo DP, Cady J, Harms MB, Baloh RH, Vandenberg SR, Yeo GW, Fu XD, Bennett CF, Cleveland DW, Ravits J, 2013. Targeted degradation of sense and antisense C9orf72 RNA foci as therapy for ALS and frontotemporal degeneration. Proc Natl Acad Sci U S A 110(47), E4530–4539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lattante S, Ciura S, Rouleau GA, Kabashi E, 2015. Defining the genetic connection linking amyotrophic lateral sclerosis (ALS) with frontotemporal dementia (FTD). Trends Genet 31(5), 263–273. [DOI] [PubMed] [Google Scholar]
- LeBel CP, Bondy SC, 1992. Oxidative damage and cerebral aging. Prog Neurobiol 38(6), 601–609. [DOI] [PubMed] [Google Scholar]
- Leslie K, Lang C, Devgan G, Azare J, Berishaj M, Gerald W, Kim YB, Paz K, Darnell JE, Albanese C, Sakamaki T, Pestell R, Bromberg J, 2006. Cyclin D1 is transcriptionally regulated by and required for transformation by activated signal transducer and activator of transcription 3. Cancer Res 66(5), 2544–2552. [DOI] [PubMed] [Google Scholar]
- Lim AC, Qi RZ, 2003. Cyclin-dependent kinases in neural development and degeneration. J Alzheimers Dis 5(4), 329–335. [DOI] [PubMed] [Google Scholar]
- Ling SC, Polymenidou M, Cleveland DW, 2013. Converging mechanisms in ALS and FTD: disrupted RNA and protein homeostasis. Neuron 79(3), 416–438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maccioni RB, Munoz JP, Barbeito L, 2001. The molecular bases of Alzheimer’s disease and other neurodegenerative disorders. Arch Med Res 32(5), 367–381. [DOI] [PubMed] [Google Scholar]
- Mehta P, Kaye W, Bryan L, Larson T, Copeland T, Wu J, Muravov O, Horton K, 2016. Prevalence of Amyotrophic Lateral Sclerosis - United States, 2012–2013. MMWR Surveill Summ 65(8), 1–12. [DOI] [PubMed] [Google Scholar]
- Mizielinska S, Lashley T, Norona FE, Clayton EL, Ridler CE, Fratta P, Isaacs AM, 2013. C9orf72 frontotemporal lobar degeneration is characterised by frequent neuronal sense and antisense RNA foci. Acta Neuropathol 126(6), 845–857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mori K, Arzberger T, Grasser FA, Gijselinck I, May S, Rentzsch K, Weng SM, Schludi MH, van der Zee J, Cruts M, Van Broeckhoven C, Kremmer E, Kretzschmar HA, Haass C, Edbauer D, 2013a. Bidirectional transcripts of the expanded C9orf72 hexanucleotide repeat are translated into aggregating dipeptide repeat proteins. Acta Neuropathol 126(6), 881–893. [DOI] [PubMed] [Google Scholar]
- Mori K, Weng SM, Arzberger T, May S, Rentzsch K, Kremmer E, Schmid B, Kretzschmar HA, Cruts M, Van Broeckhoven C, Haass C, Edbauer D, 2013b. The C9orf72 GGGGCC repeat is translated into aggregating dipeptide-repeat proteins in FTLD/ALS. Science 339(6125), 1335–1338. [DOI] [PubMed] [Google Scholar]
- Mori S, Maher P, Conti B, 2016. Neuroimmunology of the Interleukins 13 and 4. Brain Sci 6(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Musi N, Valentine JM, Sickora KR, Baeuerle E, Thompson CS, Shen Q, Orr ME, 2018. Tau protein aggregation is associated with cellular senescence in the brain. Aging Cell 17(6), e12840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Myrianthopoulos V, Evangelou K, Vasileiou PVS, Cooks T, Vassilakopoulos TP, Pangalis GA, Kouloukoussa M, Kittas C, Georgakilas AG, Gorgoulis VG, 2019. Senescence and senotherapeutics: a new field in cancer therapy. Pharmacol Ther 193, 31–49. [DOI] [PubMed] [Google Scholar]
- Ng AS, Rademakers R, Miller BL, 2015. Frontotemporal dementia: a bridge between dementia and neuromuscular disease. Ann N Y Acad Sci 1338, 71–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Noda M, Doi Y, Liang J, Kawanokuchi J, Sonobe Y, Takeuchi H, Mizuno T, Suzumura A, 2011. Fractalkine attenuates excito-neurotoxicity via microglial clearance of damaged neurons and antioxidant enzyme heme oxygenase-1 expression. J Biol Chem 286(3), 2308–2319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Norambuena A, Wallrabe H, McMahon L, Silva A, Swanson E, Khan SS, Baerthlein D, Kodis E, Oddo S, Mandell JW, Bloom GS, 2017. mTOR and neuronal cell cycle reentry: How impaired brain insulin signaling promotes Alzheimer’s disease. Alzheimers Dement 13(2), 152–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Connor SM, Stenger DA, Shaffer KM, Ma W, 2001. Survival and neurite outgrowth of rat cortical neurons in three-dimensional agarose and collagen gel matrices. Neurosci Lett 304(3), 189–193. [DOI] [PubMed] [Google Scholar]
- Oost W, Talma N, Meilof JF, Laman JD, 2018. Targeting senescence to delay progression of multiple sclerosis. J Mol Med (Berl) 96(11), 1153–1166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park J, Wetzel I, Marriott I, Dreau D, D’Avanzo C, Kim DY, Tanzi RE, Cho H, 2018. A 3D human triculture system modeling neurodegeneration and neuroinflammation in Alzheimer’s disease. Nat Neurosci 21(7), 941–951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pelegri C, Duran-Vilaregut J, del Valle J, Crespo-Biel N, Ferrer I, Pallas M, Camins A, Vilaplana J, 2008. Cell cycle activation in striatal neurons from Huntington’s disease patients and rats treated with 3-nitropropionic acid. Int J Dev Neurosci 26(7), 665–671. [DOI] [PubMed] [Google Scholar]
- Piechota M, Sunderland P, Wysocka A, Nalberczak M, Sliwinska MA, Radwanska K, Sikora E, 2016. Is senescence-associated beta-galactosidase a marker of neuronal senescence? Oncotarget 7(49), 81099–81109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ranganathan S, Bowser R, 2003. Alterations in G(1) to S phase cell-cycle regulators during amyotrophic lateral sclerosis. Am J Pathol 162(3), 823–835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ranganathan S, Bowser R, 2010. p53 and Cell Cycle Proteins Participate in Spinal Motor Neuron Cell Death in ALS. Open Pathol J 4, 11–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rashidian J, Iyirhiaro G, Aleyasin H, Rios M, Vincent I, Callaghan S, Bland RJ, Slack RS, During MJ, Park DS, 2005. Multiple cyclin-dependent kinases signals are critical mediators of ischemia/hypoxic neuronal death in vitro and in vivo. Proc Natl Acad Sci U S A 102(39), 14080–14085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rashidian J, Iyirhiaro GO, Park DS, 2007. Cell cycle machinery and stroke. Biochim Biophys Acta 1772(4), 484–493. [DOI] [PubMed] [Google Scholar]
- Rueden CT, Schindelin J, Hiner MC, DeZonia BE, Walter AE, Arena ET, Eliceiri KW, 2017. ImageJ2: ImageJ for the next generation of scientific image data. BMC Bioinformatics 18(1), 529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rutherford NJ, Heckman MG, Dejesus-Hernandez M, Baker MC, Soto-Ortolaza AI, Rayaprolu S, Stewart H, Finger E, Volkening K, Seeley WW, Hatanpaa KJ, Lomen-Hoerth C, Kertesz A, Bigio EH, Lippa C, Knopman DS, Kretzschmar HA, Neumann M, Caselli RJ, White CL 3rd, Mackenzie IR, Petersen RC, Strong MJ, Miller BL, Boeve BF, Uitti RJ, Boylan KB, Wszolek ZK, Graff-Radford NR, Dickson DW, Ross OA, Rademakers R, 2012. Length of normal alleles of C9ORF72 GGGGCC repeat do not influence disease phenotype. Neurobiol Aging 33(12), 2950 e2955–2957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seward ME, Swanson E, Norambuena A, Reimann A, Cochran JN, Li R, Roberson ED, Bloom GS, 2013. Amyloid-beta signals through tau to drive ectopic neuronal cell cycle re-entry in Alzheimer’s disease. J Cell Sci 126(Pt 5), 1278–1286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sokolowski JD, Chabanon-Hicks CN, Han CZ, Heffron DS, Mandell JW, 2014. Fractalkine is a “find-me” signal released by neurons undergoing ethanol-induced apoptosis. Front Cell Neurosci 8, 360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun Y, Coppe JP, Lam EW, 2018. Cellular Senescence: The Sought or the Unwanted? Trends Mol Med 24(10), 871–885. [DOI] [PubMed] [Google Scholar]
- Takada LT, 2015. The Genetics of Monogenic Frontotemporal Dementia. Dement Neuropsychol 9(3), 219–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tan FC, Hutchison ER, Eitan E, Mattson MP, 2014. Are there roles for brain cell senescence in aging and neurodegenerative disorders? Biogerontology 15(6), 643–660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tchkonia T, Zhu Y, van Deursen J, Campisi J, Kirkland JL, 2013. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest 123(3), 966–972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tian DS, Peng J, Murugan M, Feng LJ, Liu JL, Eyo UB, Zhou LJ, Mogilevsky R, Wang W, Wu LJ, 2017. Chemokine CCL2-CCR2 Signaling Induces Neuronal Cell Death via STAT3 Activation and IL-1beta Production after Status Epilepticus. J Neurosci 37(33), 7878–7892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van der Zee J, Gijselinck I, Dillen L, Van Langenhove T, Theuns J, Engelborghs S, Philtjens S, Vandenbulcke M, Sleegers K, Sieben A, Baumer V, Maes G, Corsmit E, Borroni B, Padovani A, Archetti S, Perneczky R, Diehl-Schmid J, de Mendonca A, Miltenberger-Miltenyi G, Pereira S, Pimentel J, Nacmias B, Bagnoli S, Sorbi S, Graff C, Chiang HH, Westerlund M, Sanchez-Valle R, Llado A, Gelpi E, Santana I, Almeida MR, Santiago B, Frisoni G, Zanetti O, Bonvicini C, Synofzik M, Maetzler W, Vom Hagen JM, Schols L, Heneka MT, Jessen F, Matej R, Parobkova E, Kovacs GG, Strobel T, Sarafov S, Tournev I, Jordanova A, Danek A, Arzberger T, Fabrizi GM, Testi S, Salmon E, Santens P, Martin JJ, Cras P, Vandenberghe R, De Deyn PP, Cruts M, Van Broeckhoven C, van der Zee J, Gijselinck I, Dillen L, Van Langenhove T, Theuns J, Philtjens S, Sleegers K, Baumer V, Maes G, Corsmit E, Cruts M, Van Broeckhoven C, van der Zee J, Gijselinck I, Dillen L, Van Langenhove T, Philtjens S, Theuns J, Sleegers K, Baumer V, Maes G, Cruts M, Van Broeckhoven C, Engelborghs S, De Deyn PP, Cras P, Engelborghs S, De Deyn PP, Vandenbulcke M, Vandenbulcke M, Borroni B, Padovani A, Archetti S, Perneczky R, Diehl-Schmid J, Synofzik M, Maetzler W, Muller Vom Hagen J, Schols L, Synofzik M, Maetzler W, Muller Vom Hagen J, Schols L, Heneka MT, Jessen F, Ramirez A, Kurzwelly D, Sachtleben C, Mairer W, de Mendonca A, Miltenberger-Miltenyi G, Pereira S, Firmo C, Pimentel J, Sanchez-Valle R, Llado A, Antonell A, Molinuevo J, Gelpi E, Graff C, Chiang HH, Westerlund M, Graff C, Kinhult Stahlbom A, Thonberg H, Nennesmo I, Borjesson-Hanson A, Nacmias B, Bagnoli S, Sorbi S, Bessi V, Piaceri I, Santana I, Santiago B, Santana I, Helena Ribeiro M, Rosario Almeida M, Oliveira C, Massano J, Garret C, Pires P, Frisoni G, Zanetti O, Bonvicini C, Sarafov S, Tournev I, Jordanova A, Tournev I, Kovacs GG, Strobel T, Heneka MT, Jessen F, Ramirez A, Kurzwelly D, Sachtleben C, Mairer W, Jessen F, Matej R, Parobkova E, Danel A, Arzberger T, Maria Fabrizi G, Testi S, Ferrari S, Cavallaro T, Salmon E, Santens P, Cras P, European Early-Onset Dementia C, 2013. A pan-European study of the C9orf72 repeat associated with FTLD: geographic prevalence, genomic instability, and intermediate repeats. Hum Mutat 34(2), 363–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Varvel NH, Bhaskar K, Patil AR, Pimplikar SW, Herrup K, Lamb BT, 2008. Abeta oligomers induce neuronal cell cycle events in Alzheimer’s disease. J Neurosci 28(43), 10786–10793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wagner L, Rechtman L, Jordan H, Ritsick M, Sanchez M, Sorenson E, Kaye W, 2015. State and metropolitan area-based amyotrophic lateral sclerosis (ALS) surveillance. Amyotroph Lateral Scler Frontotemporal Degener 17(1–2), 128–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walton CC, Zhang W, Patino-Parrado I, Barrio-Alonso E, Garrido JJ, Frade JM, 2019. Primary neurons can enter M-phase. Sci Rep 9(1), 4594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wen X, Tan W, Westergard T, Krishnamurthy K, Markandaiah SS, Shi Y, Lin S, Shneider NA, Monaghan J, Pandey UB, Pasinelli P, Ichida JK, Trotti D, 2014. Antisense proline-arginine RAN dipeptides linked to C9ORF72-ALS/FTD form toxic nuclear aggregates that initiate in vitro and in vivo neuronal death. Neuron 84(6), 1213–1225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu M, Pirtskhalava T, Farr JN, Weigand BM, Palmer AK, Weivoda MM, Inman CL, Ogrodnik MB, Hachfeld CM, Fraser DG, Onken JL, Johnson KO, Verzosa GC, Langhi LGP, Weigl M, Giorgadze N, LeBrasseur NK, Miller JD, Jurk D, Singh RJ, Allison DB, Ejima K, Hubbard GB, Ikeno Y, Cubro H, Garovic VD, Hou X, Weroha SJ, Robbins PD, Niedernhofer LJ, Khosla S, Tchkonia T, Kirkland JL, 2018. Senolytics improve physical function and increase lifespan in old age. Nat Med 24(8), 1246–1256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu X, Lei Y, Luo J, Wang J, Zhang S, Yang XJ, Sun M, Nuwaysir E, Fan G, Zhao J, Lei L, Zhong Z, 2013. Prevention of beta-amyloid induced toxicity in human iPS cell-derived neurons by inhibition of Cyclin-dependent kinases and associated cell cycle events. Stem Cell Res 10(2), 213–227. [DOI] [PubMed] [Google Scholar]
- Yoo SJ, Kim J, Lee CS, Nam Y, 2011. Simple and novel three dimensional neuronal cell culture using a micro mesh scaffold. Exp Neurobiol 20(2), 110–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang K, Donnelly CJ, Haeusler AR, Grima JC, Machamer JB, Steinwald P, Daley EL, Miller SJ, Cunningham KM, Vidensky S, Gupta S, Thomas MA, Hong I, Chiu SL, Huganir RL, Ostrow LW, Matunis MJ, Wang J, Sattler R, Lloyd TE, Rothstein JD, 2015. The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature 525(7567), 56–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu X, Raina AK, Perry G, Smith MA, 2004. Alzheimer’s disease: the two-hit hypothesis. Lancet Neurol 3(4), 219–226. [DOI] [PubMed] [Google Scholar]
- Zu T, Liu Y, Banez-Coronel M, Reid T, Pletnikova O, Lewis J, Miller TM, Harms MB, Falchook AE, Subramony SH, Ostrow LW, Rothstein JD, Troncoso JC, Ranum LP, 2013. RAN proteins and RNA foci from antisense transcripts in C9ORF72 ALS and frontotemporal dementia. Proc Natl Acad Sci U S A 110(51), E4968–4977. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplemental Figure 1. Panels a and b, Expression of βIII tubulin and tyrosine hydroxylase (TH) by neurotypic 9319a and C9+ 30i iPS neurons. Panel c, Alexa fluor secondary antibody only control. No primary antibodies were added. Anti-chicken IgY 488, anti-rabbit IgG 568, and anti-mouse IgG 647. Scale bar = 100 μm. Panel d, Viability assessment of neurotypic BOH1, C9+ 29i and C9+ 30i iPS neuronal cultures at 200 days post-differentiation.
Supplemental Figure 2. Venn diagram of differentially expressed cell cycle-associated genes in 29i and 30i iPS neurons.
Supplemental Table 1. C9+ cell lines used for neuronal progenitor cell derivation.
Supplemental Table 2. Antibodies used in studies.
Supplemental Table 3. Patient information.
Supplemental Table 4. Benjamini-Hochberg test on C9+ 29i and 30i p-values. Red text indicates hits from C9+ 29i PCR microarray studies. Bolded red text indicates genes that confirmed in C9+ 29i confirmation studies. Bolded black text indicates additional hits from C9+ 30i PCR microarray studies.