Genomic analysis of the molecular neuropathology of...

14
RESEARCH Open Access Genomic analysis of the molecular neuropathology of tuberous sclerosis using a human stem cell model Nils Grabole 1* , Jitao David Zhang 1 , Stefan Aigner 2 , Nadine Ruderisch 3 , Veronica Costa 3 , Felix C. Weber 1 , Michel Theron 1 , Nikolaos Berntenis 1 , Olivia Spleiss 1 , Martin Ebeling 1 , Gene W. Yeo 2 , Ravi Jagasia 3 and Anna Kiialainen 1 Abstract Background: Tuberous sclerosis complex (TSC) is a genetic disease characterized by benign tumor growths in multiple organs and neurological symptoms induced by mTOR hyperfunction. Because the molecular pathology is highly complex and the etiology poorly understood, we employed a defined human neuronal model with a single mTOR activating mutation to dissect the disease-relevant molecular responses driving the neuropathology and suggest new targets for treatment. Methods: We investigate the disease phenotype of TSC by neural differentiation of a human stem cell model that had been deleted for TSC2 by genome editing. Comprehensive genomic analysis was performed by RNA sequencing and ribosome profiling to obtain a detailed genome-wide description of alterations on both the transcriptional and translational level. The molecular effect of mTOR inhibitors used in the clinic was monitored and comparison to published data from patient biopsies and mouse models highlights key pathogenic processes. Results: TSC2-deficient neural stem cells showed severely reduced neuronal maturation and characteristics of astrogliosis instead. Transcriptome analysis indicated an active inflammatory response and increased metabolic activity, whereas at the level of translation ribosomal transcripts showed a 5UTR motif-mediated increase in ribosome occupancy. Further, we observed enhanced protein synthesis rates of angiogenic growth factors. Treatment with mTOR inhibitors corrected translational alterations but transcriptional dysfunction persisted. Conclusions: Our results extend the understanding of the molecular pathophysiology of TSC brain lesions, and suggest phenotype-tailored pharmacological treatment strategies. Keywords: Astrocytes, mTOR, Stem cell disease modeling, TSC2, Tuberous sclerosis Background The mTOR pathway is involved in a plethora of patholo- gies, in particular carcinogenesis [1]. However, the large number of processes downstream of mTOR and result- ing heterogeneity of clinical manifestations impede a deeper understanding of the disease mechanisms and se- lection of the optimal treatment strategy. For example, patients with tuberous sclerosis complex (TSC), caused by mutations in the tumor suppressor genes TSC1 or TSC2 leading to mTOR hyperfunction, show heterogen- eity of benign tumors and cellular dysplasia in multiple organs, including astrocytomas and cortical tubers in the brain [24]. Loss of heterozygosity for either TSC gene due to somatic mutation of the functional allele in het- erozygous patients was detected in these lesions and in- duces cancerous growth [57]. In addition, TSC patients develop central nervous system abnormalities, including structural alterations of the cortex, epilepsy, and psychiatric symptoms [8]. Clinical trials with mTOR inhibitors are on- going to treat the manifestations of this disease [9, 10]. However, while mTOR inhibitors have tremendous poten- tial as disease modifying agents, it remains unclear if they * Correspondence: [email protected] 1 Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland Full list of author information is available at the end of the article © 2016 The Author(s). Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated. Grabole et al. Genome Medicine (2016) 8:94 DOI 10.1186/s13073-016-0347-3

Transcript of Genomic analysis of the molecular neuropathology of...

Page 1: Genomic analysis of the molecular neuropathology of ...yeolab.github.io/papers/2016/Grabole_GenomeMedicine_2016.pdf · Genomic analysis of the molecular neuropathology of tuberous

RESEARCH Open Access

Genomic analysis of the molecularneuropathology of tuberous sclerosis usinga human stem cell modelNils Grabole1*, Jitao David Zhang1, Stefan Aigner2, Nadine Ruderisch3, Veronica Costa3, Felix C. Weber1, Michel Theron1,Nikolaos Berntenis1, Olivia Spleiss1, Martin Ebeling1, Gene W. Yeo2, Ravi Jagasia3 and Anna Kiialainen1

Abstract

Background: Tuberous sclerosis complex (TSC) is a genetic disease characterized by benign tumor growths inmultiple organs and neurological symptoms induced by mTOR hyperfunction. Because the molecular pathology ishighly complex and the etiology poorly understood, we employed a defined human neuronal model with a singlemTOR activating mutation to dissect the disease-relevant molecular responses driving the neuropathology andsuggest new targets for treatment.

Methods: We investigate the disease phenotype of TSC by neural differentiation of a human stem cell model that hadbeen deleted for TSC2 by genome editing. Comprehensive genomic analysis was performed by RNA sequencing andribosome profiling to obtain a detailed genome-wide description of alterations on both the transcriptional andtranslational level. The molecular effect of mTOR inhibitors used in the clinic was monitored and comparison topublished data from patient biopsies and mouse models highlights key pathogenic processes.

Results: TSC2-deficient neural stem cells showed severely reduced neuronal maturation and characteristics ofastrogliosis instead. Transcriptome analysis indicated an active inflammatory response and increased metabolic activity,whereas at the level of translation ribosomal transcripts showed a 5’UTR motif-mediated increase in ribosomeoccupancy. Further, we observed enhanced protein synthesis rates of angiogenic growth factors. Treatment withmTOR inhibitors corrected translational alterations but transcriptional dysfunction persisted.

Conclusions: Our results extend the understanding of the molecular pathophysiology of TSC brain lesions, andsuggest phenotype-tailored pharmacological treatment strategies.

Keywords: Astrocytes, mTOR, Stem cell disease modeling, TSC2, Tuberous sclerosis

BackgroundThe mTOR pathway is involved in a plethora of patholo-gies, in particular carcinogenesis [1]. However, the largenumber of processes downstream of mTOR and result-ing heterogeneity of clinical manifestations impede adeeper understanding of the disease mechanisms and se-lection of the optimal treatment strategy. For example,patients with tuberous sclerosis complex (TSC), causedby mutations in the tumor suppressor genes TSC1 or

TSC2 leading to mTOR hyperfunction, show heterogen-eity of benign tumors and cellular dysplasia in multipleorgans, including astrocytomas and cortical tubers in thebrain [2–4]. Loss of heterozygosity for either TSC genedue to somatic mutation of the functional allele in het-erozygous patients was detected in these lesions and in-duces cancerous growth [5–7]. In addition, TSC patientsdevelop central nervous system abnormalities, includingstructural alterations of the cortex, epilepsy, and psychiatricsymptoms [8]. Clinical trials with mTOR inhibitors are on-going to treat the manifestations of this disease [9, 10].However, while mTOR inhibitors have tremendous poten-tial as disease modifying agents, it remains unclear if they

* Correspondence: [email protected] Pharma Research and Early Development, Pharmaceutical Sciences,Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse124, Basel 4070, SwitzerlandFull list of author information is available at the end of the article

© 2016 The Author(s). Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, andreproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link tothe Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Grabole et al. Genome Medicine (2016) 8:94 DOI 10.1186/s13073-016-0347-3

Page 2: Genomic analysis of the molecular neuropathology of ...yeolab.github.io/papers/2016/Grabole_GenomeMedicine_2016.pdf · Genomic analysis of the molecular neuropathology of tuberous

can be effective to treat the full spectrum of TSC-associated pathophysiology.Work on mouse models identified neural progenitor

cells as the origin of brain lesions [11–15]. Nonetheless,the paucity of human cellular models has limited a bet-ter mechanistic understanding of brain lesions in TSCpatients. Hence, availability of a human TSC in vitro sys-tem to model the in vivo pathogenesis and perform ex-perimental analysis would enable discovery of noveltargets for pharmacological intervention. Recently a pio-neering study on osteosarcoma demonstrated the utilityof modeling carcinogenesis with human stem cells toelucidate disease mechanisms and identify new treat-ment options [16].Here we used human neural stem cells (NSCs) derived

from embryonic stem cells (ESCs) that have been bialleli-cally deleted for TSC2 by genome editing to study the cel-lular and molecular pathophysiology of TSC. This TSC invitro model showed reduced neuronal maturation poten-tial and increased commitment to the astrocyte lineage,providing valuable insight for the study of TSC patient bi-opsies [17]. Using RNA sequencing (RNA-Seq) and ribo-some profiling, we performed a comprehensive analysis ofthe genome-wide consequences of TSC2 loss on bothtranscription and translation. We detected a disease-relevant inflammatory response on the transcriptionallevel while translatome analysis demonstrated motif-dependent translational dysfunction of protein synthesisfactors as well as increased production of angiogenicgrowth factors. Inhibition of mTOR signaling correctedthe translation defects but not the inflammatory or angio-genic growth factor response, which were due to alteredtranscription. Thus we provide important insight into themolecular pathology of tuberous sclerosis and present anexperimental system for future investigation of disease-modifying compounds beyond mTOR inhibitors and de-velopment of comprehensive therapies for TSC.

MethodsCell line generation and neural differentiationAn allelic deletion series of TSC2 was established fromthe parental ESC line SA001 (NIH registration number0085) by use of zinc finger nucleases targeting exon 11of the TSC2 locus. Site-specific integration was con-firmed by polymerase chain reaction (PCR) amplificationof the genomic locus followed by direct sequencing. Ab-sence of non-specific integration sites was determinedby targeted locus amplification followed by deep sequen-cing. Neural conversion of ESCs to NSCs was performedusing a dual SMAD inhibition protocol. Generation ofcell lines is described and documented in detail by Costaet al. [18].NSCs were cultured according to standard methods. All

used tissue culture dishes were coated with poly-L-

ornithine (Sigma Aldrich) and laminin (Roche) and undif-ferentiated cultures were maintained in a basic mediumcomposed of a 1:1 mix of DMEM:F12 Glutamax mediumand Neurobasal medium (both Gibco, Invitrogen) thatwas supplemented with 1× B27, 1× N2, and 0.1 mM beta-mercaptoethanol (all Gibco, Invitrogen). For self-renewingconditions the following growth factors were added:10 ng/mL FGF2, 20 ng/mL BDNF (both Peprotech), and10 ng/mL EGF (R&D Systems). Ventralization was in-duced for a period of seven days by replating the cells at adensity of 12,000 cells/cm2 and changing the supplement-ing growth factors to 200 ng/mL Shh, 100 ng/mL FGF8(both Peprotech), and 100 μM ascorbic acid phosphate(Sigma Aldrich). Neuronal differentiation was initiated byreplating the cells at a density of 40,000 cells/cm2 in basicmedium supplemented with 20 ng/mL BDNF, 10 ng/mLGDNF (both Peprotech), 0.5 mM cAMP (BIOLOG LifeScience), and 100 μM ascorbic acid phosphate (SigmaAldrich). Medium was changed twice per week until theday of analysis.

Library preparation and sequencingRibosome profiling and RNA sequencing libraries wereprepared using the TruSeq Ribo Profile kit (Illumina,#RPHMR12126) as detailed in the manufacturer’s proto-col. Cells for each biological replicate of control, hetero-zygous, and homozygous cells with or without drugtreatment were washed with ice cold PBS and lysed onice in the presence of 100 μg/mL cycloheximide (Sigma).The cleared lysate was flash frozen and stored at −80 °Cuntil further processing. For ribosomal RNA depletion,the RiboZero magnetic Gold kit (Illumina) was used.Quality of amplified libraries was accessed by capillaryelectrophoresis with a high sensitivity DNA chip on a2100 Bioanalyzer (Agilent Technologies) and quantifiedby quantitative PCR with a sequencing library quantifi-cation kit (KAPA Biosystems) on a Roche Light Cycler480. Multiplexed libraries with 1 % spiked in PhiX con-trol were sequenced on a HiSeq2500 instrument for50 cycles using version 4 chemistry reagents (Illumina).BCL files were converted to the fastq format for furtherbioinformatics processing.The sequencing data from this publication is available

at the GEO database.

Bioinformatic analysisLinker tags were removed from RNA-Seq and ribosomeprofiling reads by the FASTX Toolkit, v0.0.13 (http://hannonlab.cshl.edu/fastx_toolkit/). All reads thatmapped to rRNAs, tRNAs, or mitochondrial rRNAswere removed, and the remaining reads were mapped toRefSeq (v38) by TopHat v2.0.13 [19]. Finally, all readcounts that mapped uniquely to genes were extracted

Grabole et al. Genome Medicine (2016) 8:94 Page 2 of 14

Page 3: Genomic analysis of the molecular neuropathology of ...yeolab.github.io/papers/2016/Grabole_GenomeMedicine_2016.pdf · Genomic analysis of the molecular neuropathology of tuberous

for expression analysis with the help of samtools, v1.1[20].We applied the edgeR [21] algorithm for differential gene

expression analysis and identified genes with absolute log2fold change larger than 1 and Benjamini–Hochbergadjusted p value smaller than 0.05 as significantly changed.We applied the camera [22] algorithm for gene set enrich-ment analysis with gene sets collected in the Roche internaldatabase RONET. The same method was applied forupstream regulator analysis, with transcriptional targets ofhuman transcription factors and gene expression modula-tors curated from literature in RONET as input. Geneontology enrichment analysis was performed using theFisher’s exact test.To validate our findings with results of previous stud-

ies, we retrieved gene expression data from GEO(GSE35338 for Zamanian et al. and GSE16969 for Boeret al.) or ArrayExpress (E-MEXP-2351 for Tyburczyet al.) and performed differential expression analysiswith the limma [23] package. Gene set enrichment ana-lysis was performed in the same way as RNA-Seq data toallow fair comparisons.We constructed an analysis pipeline to analyze the ribo-

some profiling data. In essence, the pipeline estimatesboth amplitude and statistical significance of differentialtranslation efficiency (TE). TE is defined as the ratiobetween expression levels measured by messenger RNAsequencing and by ribosome sequencing, with pseudo-counts of 1 added to each to avoid division by zero. Theamplitude of TE change between two conditions isestimated by the log2-transformed ratio of two TEs. Thestatistical significance is estimated by the babel method[24] based on errors-in-variables regression models.

Microfluidic quantitative PCR (qPCR)Cells from biological triplicates were harvested in TRIzolReagent (LifeTechnologies) in weekly intervals and RNAwas extracted according to the manufacturer’s instruc-tions. Traces of contaminating DNA were removedusing the TURBO DNA-free kit (LifeTechnologies). Syn-thesis of complementary DNA (cDNA) was performedusing the SuperScript III First Strand Synthesis Mix(LifeTechnologies). For the non-enzyme control, a refer-ence pool of all samples was used. cDNAs were sub-jected to specific target amplification prior to qPCRanalysis using a pool of standard predesigned TAQmanassays to be used later (Applied Biosystems) with a pre-amplification mastermix (Roche Life Science). Preampli-fied cDNA was diluted 1:5 and processed together withthe TAQman assays for analysis in 96.96 Dynamic Arrayintegrated fluidic chips on the BioMark HD platform(Fluidigm) according to the manufacturer’s instruction.Data were analyzed using Real-Time PCR Analysis soft-ware (Fluidigm).

Flow cytometryFor flow cytometry analysis, cells were analyzed as de-scribed previously [25]. In brief, cells were dissociatedwith Accutase (Sigma, A6964), treated with DNase(Sigma, D5025), and resuspended in sorting medium(growth medium supplemented with 0.5 % BSA and5 mM EDTA). 1 × 106 cells were stained with antiCD184 (BD555976) and anti CD44 (BD555479) anti-bodies according to the manufacturer’s recommenda-tions for 20 min at 4 °C. Stained cells were washed oncemore with sorting medium and centrifuged at 250 g for5 min before acquisition of data on a FACSCanto II flowcytometer. For analysis of protein synthesis rates, theClick-iT Plus OPP kit (LifeTechnologies, C10456) wasused and cells prepared according to the manufacturer’sinstructions. Data were analyzed using FlowJo v10.0.8software.

Western blot analysis and protein arraysCells for protein analysis were directly lysed using RIPAbuffer (LifeTechnologies) supplemented with proteaseand phosphatase inhibitors (Roche Life Science). Totalprotein concentration was quantified by BCA assay(LifeTechnologies). For SDS-PAGE, 20 μg of total pro-tein lysate were analyzed by western blotting on nitrocel-lulose membranes (BIO-RAD). For western blot analysis,all antibodies were used at 1:1000 (except anti-β-actinwhich was used 1:20,000) and chemiluminescence wasdetected with ECL Reagent (GE Healthcare) using a Fu-sion FX system (Vilber Lourmat). The same membranesused to detect proteins of interest were stripped,blocked, and reprobed with anti-beta-Actin antibody.Primary antibodies used were mouse-anti-STAT3 (CST#9139), rabbit-anti-phospho-STAT3 (Tyr705) (CST#9131), rabbit-anti-GFAP (Dako Z0334), HRP linkedmouse-anti-beta-Actin (Abcam ab49900), rabbit-anti-S6ribosomal protein (CST #2217), and rabbit-anti-phosphoS6 ribosomal protein (Ser240/244) (CST #2215). Sec-ondary antibodies used were HRP linked anti-mouse(Santa Cruz Biotechnology sc-2005) and HRP linkedanti-rabbit (Santa Cruz Biotechnology sc-2030). Forquantification of cytokines and angiogenesis factors, thesecreted proteins in conditioned media were detectedusing Proteome Profiler array kits (R&D Systems) ac-cording to the manufacturer’s instructions. The intensityof the signals was quantified and normalized by densi-tometry using ImageJ software.

ImmunocytochemistryFor immunofluorescence analysis, cells were cultured in96-well plates (BD Falcon), fixed in 4 % PFA in PBS for10 min, permeabilized with 0.25 % (v/v) Triton X-100 inPBS for 5 min, and incubated for 45 min at roomtemperature in blocking solution (10 % BSA w/v in

Grabole et al. Genome Medicine (2016) 8:94 Page 3 of 14

Page 4: Genomic analysis of the molecular neuropathology of ...yeolab.github.io/papers/2016/Grabole_GenomeMedicine_2016.pdf · Genomic analysis of the molecular neuropathology of tuberous

PBS). Primary (anti-GFAP DAKO, anti-HuC/D LifeTechnologies, anti-Doublecortin Santa Cruz) and sec-ondary antibodies were diluted in 3 % BSA (w/v) and in-cubated overnight at 4 °C and for 1.5 h at roomtemperature, respectively. Upon removal of secondaryantibody, cells were incubated with DAPI (1 μg/mL) inPBS for 5 min at room temperature. Confocal fluores-cence images were acquired using a Leica TCS SP5(Leica Microsystems) inverted microscope. Stacks ac-quired along the z-axis were converted into maximumprojections using LAS-AF software.

ResultsTSC2-deficient neural stem cells exhibit altered neuronaland glial differentiationLoss of heterozygosity was reported in TSC patient biop-sies and mouse models showed that brain lesions arisefrom neural progenitor cells [6, 12]. In order to investigatethe pathogenesis of these brain lesions, we employed fore-brain neural stem cells derived from human embryonicstem cells as an in vitro model. Two TSC2-deficient celllines were derived from a parental stem cell line byperforming genome editing using zinc finger nucleases todisrupt both TSC2 alleles as described by Costa et al. [18].In control cell lines, the same inactivating cassette wasinserted into the AAVS1 locus, which induces no pheno-typic effects [26], and absence of off-target integrationswas determined by targeted locus amplification sequen-cing. In contrast to the control cell lines, TSC2-deficientcells showed no detectable TSC2 protein expression andincreased phosphorylation of mTOR target ribosomalprotein S6 (RPS6), indicating mTOR hyperfunction(Additional file 1: Figure S1A).We applied a differentiation protocol that induces a

temporal order of neurogenesis and gliogenesis similarto in vivo neurodevelopment with cells acquiring expres-sion of markers of functional neural cell types as well asmature electrophysiological properties within six weeksof differentiation. Reproducibility of the protocol wasshown by the high degree of correlation of protein ex-pression changes after 41 days of differentiation acrossdifferent wild-type NSC lines with at least five independ-ently differentiated biological replicates [27]. To monitorcell fate over the course of neuronal differentiation, wecollected samples in weekly intervals (Fig. 1a) and deter-mined transcript levels of neuronal and glial differenti-ation and maturation markers by qPCR (Fig. 1b andAdditional file 1: Figure S1B). At the start of differenti-ation, TSC2-deficient cells downregulated neural stemcell marker SOX2 similar to control cells. However, atweek 3 of differentiation, pro-neural transcription factorMASH1 was expressed at significantly lower levels inTSC2-deficient cells. This suggests that generation offunctional neurons might be affected, although at the

same time induction of TBR2, associated with a neur-onal progenitor state, was observed. In contrast to neur-onal fate regulator MASH1, the glial cell fate regulatorsNF1B and NF1X were expressed earlier and stronger aswell.Eventually, at week 6, TSC2-deficient cultures exhib-

ited in comparison to controls severely reduced levels oftranscripts required for functional maturation of gluta-matergic and GABAergic neurons. These include bothpre- and postsynaptic markers, such as GABA and glu-tamate receptors, and neural cell adhesion proteins.Equally, markers of oligodendrocytes (OLIG1, OLIG2)and astrocytes (ALDH1L1, S100B) as well as other crit-ical genes for astrocyte functionality like glutamatetransporters (EAAT3, GLAST, and GLT-1) were stronglydownregulated, together indicating that TSC2 is requiredfor generation of functionally mature cell types.A reduced differentiation of radial glia cells into oligo-

dendrocytes leading to myelination defects is also seenin the TSC mouse models [28–30]. Simultaneously themajority, but not all, of these models show a strong up-regulation of GFAP as a marker of immature glia andastrogliosis [28, 29, 31], which can also be found in TSCpatient biopsies [32]. Notably, such markers (GFAP,AQP4) also showed stronger expression in our TSC2-de-ficient cells (Fig. 1b), suggesting an altered cell fate alsoin the in vitro model. This was also supported by flowcytometry analysis of CD44 and CD184 (Additional file1: Figure S1C), which can monitor the generation ofneuronal and glial populations from human pluripotentcells [25]. Therefore, we confirmed GFAP expression atthe protein level by immunofluorescence staining, re-vealing a stark increase in the number of cells expressingGFAP (Fig. 1c and Additional file 1: Figure S1D). Fur-thermore, western blot analysis showed that there was asignificant increase in GFAP protein levels in TSC2-defi-cient cultures (Fig. 1d). As STAT3 signaling is linked toGFAP expression and involved in regulation of astrocytedevelopment and phenotype [33, 34], we determinedSTAT3 activity by western blot analysis and detected asignificant increase in phospho-STAT3 levels in ourTSC2-deficient cell lines (Fig. 1e).In summary, using our human cellular model, we

showed that differentiating TSC2-deficient NSCs exhibitincreased expression of markers indicative of an earlyastroglial lineage and reduced differentiation in matureneuronal and astroglial cell types.

Transcriptome analysis illustrates an active inflammatoryresponse in the TSC2 brain lesion modelTo characterize the aberrant cell state due to absence ofTSC2, we performed RNA-Seq on the cells after sixweeks of differentiation. We included two TSC2

Grabole et al. Genome Medicine (2016) 8:94 Page 4 of 14

Page 5: Genomic analysis of the molecular neuropathology of ...yeolab.github.io/papers/2016/Grabole_GenomeMedicine_2016.pdf · Genomic analysis of the molecular neuropathology of tuberous

a

dc

b

e

TSC2-/-

mer

ge

Control

DC

X

GF

AP

TSC2-/- (T1) Control TSC2-/- (T2)

Rel

ativ

e m

RN

A e

xpre

ssio

n

GABAergic neuron markers

***

-1 0 1 2 3 4 5 6

AQP4

-1 0 1 2 3 4 5 6

GFAP

-1 0 1 2 3 4 5 6

ALDH1L1

Astrocyte markers

***

*

Glutamatergic neuron markers

*** **

**

Astrogenesis markers

****

***

**

Neurogenesis markers

Glutamate transporter

-1 0 1 2 3 4 5 6

GLAST (SLC1A3)

-1 0 1 2 3 4 5 6

GLT-1 (SLC1A2)

-1 0 1 2 3 4 5 6

EAAT3 (SLC1A1)

***

**

200 µm

GFAP

ACTIN

C1 C1’ C2’C2Control TSC2 -/-

T1 T1’ T2’T2

0,0

0,5

1,0

1,5

2,0

2,5

rela

tive

GF

AP

leve

l

***

0,0

0,5

1,0

1,5

2,0

2,5

rela

tive

p-S

TA

T3

leve

l

*

STAT3

pSTAT3

C1 C2 T2T1Control TSC2 -/-

C1 C2 T2T1Control TSC2 -/-

C1 C1’ C2’C2Control TSC2 -/-

T1 T1’ T2’T2

qPCR

NSCsWeek -1 Week 0 Week1 Week2 Week3 Week4 Week5 Week6

PatterningRNAseq

Western Blot

week of differentation

Fig. 1 Reduced functional maturation in the absence of TSC2. a Experimental protocol outline. For an experiment neural stem cells, generally maintained inself-renewing conditions (week −1), were subjected to a patterning step for seven days (week 0) before induction of neural differentiation (week 1–6).Samples were collected in weekly intervals for gene expression quantification by qPCR. After six weeks of differentiation, additional samples were collectedfor western blot, immunofluorescence, RNA-Seq, and ribosome profiling. b Relative quantification of expression levels of the indicated neural marker genesover the course of six weeks of differentiation in control and TSC2 deleted cell lines. Expression levels in control cells after six weeks of differentiation arenormalized to 1. Error bars show standard error of the mean and asterisks indicate significant difference in a two-tailed Student’s t-test, *p < 0.05, **p < 0.01,***p < 0.001. c Representative confocal images of 6-week-old neural cultures of control and TSC2 deleted cell lines stained for doublecortin (DCX) andGFAP. Confocal z-axis stacks were acquired and restructured. d, e Western blot analysis for the indicated proteins of neural cultures after six weeks ofdifferentiation using independent derived control (C1, C2) and TSC2 deletion (T1, T2) cell lines in biological replicates (e.g. C1 and C1’). Densitometricanalysis of GFAP protein and STAT3 phosphorylation levels is normalized to beta Actin and total STAT3 levels, respectively. Data are normalized to the levelin control cell line C1 and error bars represent standard deviation. Significance is determined by Student’s two-tailed t-test, *p < 0.05, ***p < 0.001

Grabole et al. Genome Medicine (2016) 8:94 Page 5 of 14

Page 6: Genomic analysis of the molecular neuropathology of ...yeolab.github.io/papers/2016/Grabole_GenomeMedicine_2016.pdf · Genomic analysis of the molecular neuropathology of tuberous

heterozygous cell lines derived from the same parental linein the analysis to serve as cellular models recapitulatingheterozygous tissue of TSC patients and to study TSC2gene dosage dependent effects. While only ten genesshowed significant expression changes in TSC2 heterozy-gous cells, there were over 2000 genes either upregulatedor downregulated in TSC2 depleted cells (Additional file2: Figure S2A and Additional file 3: Table S1). Corres-pondence analysis of transcriptomes, which is concep-tually similar to principal component analysis, yetmore suitable for discrete sequencing counts [35], il-lustrated the similarity between control and heterozy-gous cells as well as the dissimilarity of those toTSC2 homozygous cells (Fig. 2a).We aimed to further explore the astrocyte phenotype

in TSC2-deficient cultures that was suggested by the al-tered expression of astrocytic markers. Therefore, wecompared our RNA-Seq data to published gene expres-sion data of mouse models of astrogliosis, which showstrong activation of astrocytes [36]. There was a highlysignificant overlap of misregulated genes among thedatasets as determined by Fisher’s exact test (p = 4.9e-25and 5.5e-10, respectively), despite the overall lower num-ber of genes identified by microarray (Fig. 2b). Gene setenrichment analysis (Additional file 2: Figure S2B–F)identified many induced genes to be implicated in meta-bolic pathways. In particular the enzymes of almost allcatalytic steps in the glycolytic pathway were induced,highlighting an increased metabolic activity in the ab-sence of TSC2 (Fig. 2c). Moreover, the enrichment ininflammation-related functional categories (Additionalfile 4: Table S2) supports the notion of an altered astro-cyte state.To identify activation of which signaling pathways

could induce the transcriptional changes observed, weperformed upstream regulator analysis with ourcomplete dataset. In particular, inflammatory cytokinesignaling was highlighted, for example by interleukinsand interferons (Fig. 2d); together with transcription fac-tors, such as AP1 and NFkB, which are key mediators ofthe transcriptional response to inflammation. AlsoSTAT3 activation was identified here again, consistentwith the higher STAT3 phosphorylation levels detectedin western blot analysis. Other interesting pathways withsignificant enrichment were related to hypoxia and an-giogenic growth factors like VEGF and PDGF.We addressed the clinical relevance of these findings

by comparison with published microarray data fromresected cortical tubers of TSC patients [37]. Notably,there was a strong correlation in gene ontology term en-richments for our in vitro model with those for the biop-sies (Fig. 2e and Additional file 5: Table S3).Inflammation-related terms showed enrichment in bothdatasets, such as immune response with a p value of

0.0028 for patient tubers and 0.00014 for the in vitromodel, thereby attesting relevance to our findings.Shared upregulated genes included interferon-induciblegenes, such as GBP1 and GBP2. Also, genes implicatedin regulation of blood vessel development, such as mem-bers of the angiopoietin family, were induced in bothtypes of samples leading to significant enrichment of re-spective ontology terms and pointing to another patho-logically relevant process. In line with the results on cellfate changes, this global analysis shows that genes withreduced expression were enriched for terms related tosynaptic transmission in both the in vitro model and pa-tient biopsies. In addition, we compared our data topublicly available microarray data from subependymalgiant cell astrocytomas (SEGAs) that also arise in TSCpatients and show a larger contribution of glial cells ascompared to cortical tubers. The correlation in gene ex-pression changes to these glioneural tumors was evenstronger (Fig. 2f and Additional file 6: Table S4) withmore gene sets passing the significance threshold andshowing the same direction of deregulation in both invitro and in vivo samples. Notably, again inflammationand blood vessel development were the key categoriesinduced, while neuronal differentiation and related termswere those most significantly decreased. Together thisanalysis indicates that the same type of disease processesis relevant to all three sample types and that our modelalso molecularly is closer to astrocytomas than corticaltubers.Of these disease-relevant processes, neuroinflamma-

tion in particular is of interest as it is strongly linked toepileptogenesis and likely plays a role in generation ofseizures in TSC patients [38]. Thus, we sought to deter-mine levels of inflammatory markers with reportedexpression in brain lesions by using protein arrays(Additional file 2: Figure S2F). We detected a 12-foldinduction of the soluble form of ICAM1, an important in-dicator for activity of the proinflammatory cascade in as-trocytes of TSC lesions [39]. Moreover, there was a nine-fold increase in plasminogen activator uPA that is stronglyinduced in epileptogenic cortical tubers [40]. Finally, therewas a more modest, yet still significant induction of IL6and IL8 reported for cortical tubers and lung cysts of TSCpatients, respectively [41, 42]. Interestingly, inflammatorysignaling was recently suggested to be involved in epilep-togenesis in a TSC1 mouse model [43], further highlight-ing the importance to study inflammatory mechanisms infuture efforts to treat TSC-associated epilepsy.Based on the similarities of physiological processes

transcriptionally altered between brain lesion biopsiesand our TSC2 deletion in vitro model, we conclude thatour model holds disease relevance and provides valuableinsight into the molecular pathology of TSC. In particu-lar, using our comprehensive dataset from the human

Grabole et al. Genome Medicine (2016) 8:94 Page 6 of 14

Page 7: Genomic analysis of the molecular neuropathology of ...yeolab.github.io/papers/2016/Grabole_GenomeMedicine_2016.pdf · Genomic analysis of the molecular neuropathology of tuberous

a

Correspondance axis 1 (58.7%)

Cor

resp

onda

nce

axis

2 (

13.1

%)

Heteroz.Control Homoz.

-0.3 -0.2 -0.1 0.0 0.1 0.2 0.3 0.4

-0.2

-0.1

0.0

0.1

0.2

b

d

TSC2 -/- in vitro model

mouse models (astrocyte activation)

824

99 47180

4314

29 P-Val : 5.5e-10P-Val : 4.9e-25

cIL1AIL6IL1BIL13IL17ATLR2TLR4IFNgIFNaJUNBJUNJUNDSPP1TNF (-)OSMCSF2NF-kBCEBPBSTAT3STAT1SMAD4SMAD3TGFB2TGFB1VEGFBPDGFBAGTHIF1BHIF1AADIPOQINSTFE3ATF4CHD7 (-)USF1

HSF1ATF2

signaling pathway

Interleukin

Toll-like receptor

Interferon

AP-1

related

JAK/STAT

TGF beta

Angiogenesis

Hypoxia

Other

log2 (fold change RNA)0 54321

GLUT1

HK2Glucose

Glucose 6-P

Fructose 6-P

Fructose 1,6-bis-P

Glyceraldehyde 3-P

1,3-Bisphosphoglycerate (BPG)

2,3-BPG

3-Phosphoglycerate

2-Phosphoglycerate

Phosphoenolpyruvate

Acetyl-CoALactate

PFKP PFKL

ALDOA

BPGM

PGK1

PGAM1 PGAM2

LDHD

GPD1GPD2

PDK1 PDK3

PKM

ENO1 ENO3

logFC(RNA)0.5 - 1

> 2

1 - 1.51.5 - 2

DHAP

Pyruvate

e

−2 0 2 4 6

−10

−5

05

Enrichment score

GO Enrichment P−value (in vivo)

GO

Enr

ichm

ent P

−va

lue

(in v

itro)

Cortical tubers; Boer et a l. (GO Enrichment P-value)

In v

itro

mo

del

(G

O E

nric

hmen

t P-v

alue

)

TSC model vs. Cortical tubers f

In v

itro

mo

del

(G

O E

nric

hmen

t P-v

alue

)

SEGA; Tyburczy et al. (GO Enrichment P-value)

TSC model vs. SEGAs

Fig. 2 Transcriptional signatures of inflammation and angiogenesis mark the molecular pathology of TSC. a Correspondence analysis of RNA-Seq resultsfrom control (blue), TSC2 heterozygous (orange), and homozygous mutant (red) cell lines after six weeks of differentiation. Each dot represents a biologicalreplicate. b Venn diagram showing overlap of genes deregulated in two mouse models of astrocyte activation (blue) [36] with those in TSC2-deficient cells(red). Significance of overlap between gene lists (p values of 4.9E-25 and 5.5E-10) was determined by Fisher’s exact test. c Pathway map for glycolysisenzymes indicating log2 fold changes of transcript levels in TSC2-deficient cells. d Whisker box plot for target gene expression changes of listed upstreamregulators/signaling pathways that were found significantly (false discovery rate (FDR) < 1E-4) and consistently over-expressed in TSC2-deficient cells. Verticallines (median); error bars (1.5 times the interquartile range); (−): negatively regulated targets. e, f Gene Ontology gene set enrichment in in vivo (x-axis) andin vitro (y-axis) samples. Gene set analysis based on Fisher’s exact test was applied to the comparison of cortical tubers versus normal cortex autopsies fromcontrols (Boer et al. [37]) or SEGAs versus periventricular control tissue from non-TSC patients against TSC2-deficient cells versus control cells. Genes that aresignificantly differentially expressed (cutoff: FDR < 0.05) are tested for over-representation of Gene Ontology biological process terms. The p values arelog10 transformed and given a sign according to the direction of gene expression changes in the absence of TSC2. Only significantly enriched terms(p < 0.05) are reported and no extra filtering was applied. All changes that we observed were consistent in both conditions. Selected points are textlabeled; numerical values and labels of all points can be found in the accompanying source data

Grabole et al. Genome Medicine (2016) 8:94 Page 7 of 14

Page 8: Genomic analysis of the molecular neuropathology of ...yeolab.github.io/papers/2016/Grabole_GenomeMedicine_2016.pdf · Genomic analysis of the molecular neuropathology of tuberous

model system, we could provide deeper insight into therelevant roles of inflammation and angiogenesis in TSCpathology.

Ribosome profiling demonstrates increased proteinsynthesis of growth factors in TSC2-deficient cellsIn addition to transcriptional changes, TSC2-deficientcells are expected to show translational changes asmTOR signaling is a key regulator of translation initi-ation [44]. To detect translational dysfunction in ourbrain lesion model after six weeks of differentiation weperformed ribosome profiling, which allows quantifica-tion of dynamic changes in translation of all transcriptsand can serve as a measure of protein synthesis on aproteome-wide scale by using deep sequencing [45]. Incells homozygous for TSC2 deletion but also in hetero-zygous cells, having shown almost no transcriptionalchanges, translationally deregulated transcripts were de-tected (Fig. 3a). That the majority of transcripts withchanged translation showed no simultaneous change intranscript abundance indicates a regulatory control oftranslation independent of transcriptional control.To explore such regulatory control, we performed motif

discovery within the 5’UTR of affected transcripts, be-cause mTOR responsiveness of transcripts can be regu-lated by presence of a terminal oligopyrimidine (TOP)motif in the 5’UTR [46]. Enrichment for such apyrimidine-rich motif could be confirmed among transla-tionally regulated transcripts in TSC2 deletion homozy-gous cells (Fig. 3b), but not in heterozygous cells. Thepresence of this motif in a transcript was associated withan increase in TE by two-fold to four-fold in TSC2-defi-cient cells, but not for the same transcripts in heterozy-gous cells (Fig. 3c). To identify overrepresented functionalcategories associated with mistranslated transcripts and tounderstand what impact the translational dysfunction mayhave on the cellular phenotype, we performed gene set en-richment analysis. While no functional category wasenriched for the set of transcripts with altered translationin heterozygous cells, a clear and translation level specificenrichment for a function related to protein synthesis wasidentified in TSC2 deletion homozygous cells (Fig. 3d,Additional files 7 and 8: Figure S3A and Table S5).To test if this induction has a direct consequence on

the biosynthetic capacity of the cells, we went on toprobe protein synthesis rates. Using flow cytometry, theincorporation rate of an amino acid analog into newlysynthesized protein was determined and we could con-firm that homozygous cells indeed showed higher syn-thesis rates than heterozygous or control cells (Fig. 3e).To elucidate what biological processes might be affectedmost we determined their respective enrichment amongproteins with a synthesis rate raised by at least four-fold.Proteins with enhanced synthesis rates were most

frequently implicated in induction and control of bloodvessel formation such as VEGF, PDGF, and AGT (Fig. 3f ).This demonstrates that the observed increased transcrip-tion of angiogenic factors, recapitulating findings frompatient biopsies, leads to higher protein synthesis rates.An additional process highlighted in this analysis wastissue remodeling, which always needs to be closelyaligned with regulation of angiogenesis, and their con-current enrichment points to a co-regulation of theselinked processes. To validate these ribosome profiling re-sults we used protein arrays to confirm higher produc-tion of angiogenic factors by TSC2-deficient cells and wewere able to confirm significantly induced levels of theangiogenic growth factors VEGF, HGF, PDGFA, ANG,and ANGPT1 (Fig. 3g and Additional file 9: Table S6).In summary, we could demonstrate a 5’UTR pyrimidine-

rich motif dependent increase in translation of factors in-volved in protein synthesis in the absence of TSC2. Suchtranslation-specific regulation during carcinogenesis waspreviously also described for transcript subsets implicatedin metastasis and regulation of oxidative stress [47, 48].Here, it illustrates the control of the mTOR pathway overthe cell’s protein synthesis capacity, a mechanism frequentlyexploited by oncogenic events driving hypertrophic growth[49, 50]. Elevated synthesis of angiogenic growth factors inour neural cell-specific in vitro model implies that mutantcells may have the ability to promote blood vessel forma-tion to sustain the growth of TSC2-deficient astrocytomas.This notion is also coherent with the highly vascularizedtumors of TSC patients in other organs, like angiofibromasand angiomyolipomas [51]. In support of the pro-angiogenic phenotype we observe in our model, it was re-cently reported that an endothelial-cell-specific deletion ofTsc1 in a mouse model of angiosarcoma led to retinalangiogenesis and formation of vascular tumors [52]. Block-ing of autocrine VEGF signaling in this mouse model wasable to abolish vascular tumor development and growth.Therefore, inhibition of angiogenesis should also be consid-ered as an alternative treatment strategy for astrocytomasrequiring surgery.

Pharmacological inhibition of mTOR corrects translationaldefects but not the pathologic cellular stateHaving recognized angiogenic growth factors, an inflam-matory response, and induced translation of the proteinsynthesis machinery as altered disease-relevant mecha-nisms in our TSC model with mTOR hyperfunction, weset out to investigate the therapeutic molecular effects ofmTOR inhibitors. To mimic treatment of establishedbrain lesions in TSC patients, we treated TSC2 deletedcells with mTOR inhibitors after six weeks of differenti-ation and performed ribosome profiling and RNA-Seqanalysis. In clinical trials rapamycin analogs were shownto induce regression of astrocytomas in TSC patients,

Grabole et al. Genome Medicine (2016) 8:94 Page 8 of 14

Page 9: Genomic analysis of the molecular neuropathology of ...yeolab.github.io/papers/2016/Grabole_GenomeMedicine_2016.pdf · Genomic analysis of the molecular neuropathology of tuberous

-log (p value)

Blood vessel development

Vasculature development

Blood vessel morphogenesis

Tissue remodeling

Extracellular matrix organization

0 2 4 6 8 10

a

c5’TOP transcripts All other transcripts

f

d

b

1 14131211109876543 1520

2

1Bit

s

En

rich

men

t sc

ore

(tr

ansl

atio

n)

Enrichment score (transcription)

Heterozygous

0 42-4 -2

0

4

2

-4

-2

Heterozygous

Protein synthesis

Homozygous

Enrichment score (transcription)

En

rich

men

t sc

ore

(tr

ansl

atio

n)

0 42-4 -2

0

4

2

-4

-2

Homozygous

e

translation only translation and transcription

TSC2 +/-TSC2 -/-

translationally regulated transcripts

Angiogenic growth factors

0

3

6

9

12

15

HGF

ANGPT1ANG

PDGF-AVEGF

Fo

ld in

du

ctio

n

*****

***

***

***

-2 210-1

% o

f to

tal

0

15

20

10

5

-2 210-1

Het

ero

z.C

on

tro

l

cell

cou

nt

-103 0 105104103

Ho

mo

z. (

T2)

Ho

mo

z. (

T1)

g

22

77

5

133

Fig. 3 TOP motif mediated enhancement of growth factor synthesis. a Pie chart representation of the number of transcripts showing a significantchange on the level of translation only (red; translation changes normalized by transcript level changes) or also on the transcriptional level (blue).b Detected de novo motif enrichment using the MEME tool (p < 0.05) within the 5’UTR of genes that showed significant TE changes inTSC2-deficient cells. c Density histograms for log2 changes of TE comparing heterozygous (left panel) and homozygous cells (right) with control.Red bars indicate transcripts with a known 5’-TOP motif and gray bars indicate all other detected transcripts. d Scatterplots of gene set enrichmentanalysis (GSEA) significance scores comparing transcriptional (x-axis) and translational (y-axis) changes in TSC2 heterozygous (left panel) andhomozygous cells (right panel) versus control. Multiple pathways associated with protein synthesis (labeled) are positively regulated specifically onthe translational level (FDR < 1E-4). e Measurement of translational activity in control, TSC2 heterozygous, and homozygous mutant cells after sixweeks of differentiation. Protein synthesis was measured by flow cytometric detection of incorporation of FITC labeled amino acid analogO-propargyl-puromycin into newly synthesized protein. Histograms display the cell count for different levels of OPP incorporation. f Network of“vasculature development” and “tissue remodeling” related transcripts that show induction (logFC ≥ 2) of translation in the absence of TSC2 asdetermined by Fisher’s exact test (FDR < 0.01). Ellipse nodes represent transcripts and edges indicate either gene-biological process associations orprotein-protein interactions. g Detection of angiogenic growth factors in cultures of TSC2-deficient and control cell lines (n=3) using proteinarrays. Expression levels in control cells are set to 1 and significance of differential expression is determined by Student’s two tailed t-test,*p <0.05, **p <0.01, ***p <0.001

Grabole et al. Genome Medicine (2016) 8:94 Page 9 of 14

Page 10: Genomic analysis of the molecular neuropathology of ...yeolab.github.io/papers/2016/Grabole_GenomeMedicine_2016.pdf · Genomic analysis of the molecular neuropathology of tuberous

which however can regrow once treatment is stopped[10, 53]. Besides rapamycin that inhibits mTOR allosteri-cally, we also tested the effect of ATP-competitivemTOR kinase inhibitor AZD-8055 [54].Consistent with the central role of mTOR in activation

of translation, treatment with both inhibitors primarilyled to reduced translation of transcripts and had aweaker effect on transcription (Fig. 4a and Additionalfile 10: Table S7). Among the transcripts showing down-regulated translation after inhibitor treatment of TSC2deleted cells, there were 76 target transcripts shared byboth rapamycin and AZD-8055. This shared set repre-sents three-quarters of all transcripts affected in eithertreatment, suggesting a consistent set of targets affectedby mTOR inhibition (Fig. 4b). Importantly, this sharedset of translationally downregulated transcripts includedmore than half of the transcripts that were translation-ally upregulated in vehicle treated TSC2 depleted cells ascompared to controls, indicating an at least partial res-cue of the molecular phenotype.In accordance with our previous results, these tran-

scripts were essentially all implicated in protein synthesisand mostly coding for ribosomal proteins. When lookingspecifically at ribosomal transcripts showing inducedtranslation in untreated TSC2-deficient cell lines, it wasobvious that both rapamycin and AZD-8055 treatmentwere able to correct elevated translation levels, implyinga reversal of the excessive protein synthesis (Fig. 4c andAdditional file 11: Table S8). In contrast, increased ex-pression of genes related to angiogenesis and inflamma-tion, both based on induced transcription with noalterations at the level of translation, remained un-affected by treatment with mTOR inhibitors. Accord-ingly, the respective protein output remained high, evenin the drug-treated cells (Fig. 4d). Along these lines, wefound that rapamycin treatment also did not reduceSTAT3 phosphorylation levels in TSC2-deficient cells(Fig. 4e). This suggests that disease mechanisms basedon transcriptional changes cannot be treated withmTOR inhibitors even though the initial manifestationof the pathophysiology was dependent on mTOR hyper-function and future work will address the feasibility andsuccess of combinatorial treatment.This analysis highlights the importance of monitoring

transcription and translation independently when inves-tigating modulation of gene expression by drug treat-ment. We could demonstrate that both inhibitors have asimilar effect and corrected excessive translation of ribo-somal proteins with possible implications for reducinghypertrophy of brain tumors in TSC patients. Nonethe-less, the transcriptional signature of an inflamed stateand angiogenesis remained despite mTOR inhibitortreatment. This shows that while mTOR was effectivelyinhibited, the cellular pathophysiology was not fully

reversed and suggests use of additional combinationtherapies tailored to the clinical picture of TSC patients(Fig. 4f ).

DiscussionApplying comprehensive genomic analysis to our humanstem cell disease model enabled us to gain a deeper un-derstanding of the TSC neuropathology. In TSC2-deficient cultures we detect more cells expressing GFAP,higher levels of STAT3 activation, and increased expres-sion of inflammation associated genes. All these featuresare indicative of aberrant glial differentiation [34, 36, 55]and are also in agreement with reports on cortical tubersand SEGAs from TSC patients [5, 17, 32, 39]. Import-antly, it is now evident that astrocyte pathophysiologycan have a detrimental impact on neuronal function aswell [56, 57]. Interestingly, an increase in GFAP express-ing astrocyte-like cells is equally observed in mousemodels for RASopathies including neurofibromatosis,Costello syndrome, Noonan syndrome, and cardio-facio-cutaneous syndrome [58–61]. This illustrates that glialdifferentiation is frequently perturbed by excessive acti-vation of growth signaling.Mechanistically of interest for astrocyte dysfunction,

appear, in particular, the processes of neuroinflammationand the altered expression of genes regulating glutamatehomoestasis. In addition, elevated angiogenic growthfactor levels in our neural model suggest enhancedvascularization of brain lesions in analogy to the highlyvascularized angiofibromas and angiomyolipomas inTSC patients [51]. As angiogenesis might be a prerequis-ite for increased synthetic capacity and hypertrophicgrowth of astrocytomas, it constitutes a possible targetfor pharmacological intervention in TSC.Interestingly, in contrast to angiogenic growth factors

as in our model of benign hypertrophic astrocytomas,previous work on malignant prostate cancer highlightedmetastasis as most significant translationally inducedprocess [47], suggesting that the rerouting of the transla-tional program upon mTOR hyperfunction can be con-text dependent.Thus, diseases that could be grouped as “mTORopa-

thies,” due to similar etiology, still present with vastly dif-ferent phenotypes that require different treatment. As weshow in our TSC model, even for a single disease withheterogeneous phenotypes, inhibition of only mTOR sig-naling may not be sufficient to treat the entire spectrumof pathology. An acquired aberrant cellular state can beindependent of mTOR signaling, therefore treatmentstrategies then need to be tailored to the respectivephenotype.Our stem cell model allows the profiling of molecular

alterations present during human embryonic develop-ment and then the tracking of aberrant cellular

Grabole et al. Genome Medicine (2016) 8:94 Page 10 of 14

Page 11: Genomic analysis of the molecular neuropathology of ...yeolab.github.io/papers/2016/Grabole_GenomeMedicine_2016.pdf · Genomic analysis of the molecular neuropathology of tuberous

a

c

b

e

0 3-3

log2(TE)C1 C2 T1 T2 T1 T1 T2 T2

control TSC2 -/-+DMSO +DMSO +Rapa +AZD +Rapa +AZD

Ribosomal proteins

DownregulatedUpregulated

Translation

AZD-805

5

Rapam

ycin

15103

29

9953

232812

AZD-805

5

Rapam

ycin

Transcription

Expression changes upon inhibitor treatment Up in TSC2 -/-

Down by Rapamycin

Down by AZD-8055

24

233

25

43

23

(DMSO treated) Down in TSC2 -/-

Up by Rapamycin

Up by AZD-8055

20

25 122

11 0

(DMSO treated)

d

no

rmal

ized

log

2(T

E)

Protein synthesis Angiogenesis

-1

1

0

-2

-3

2

3

no

rmal

ized

RN

A (

cpm

) n

orm

aliz

ed R

PF

(cp

m)

0

-5

-10

5

10

0

-5

-10

5

10

Tran

scri

pti

on

Tran

slat

ion

Pro

tein

ou

tpu

t

wt+ DM

SO

TSC2 -/-+ AZD-8055

+ Rapamycin

+ DMSO

wt+ DM

SO

TSC2 -/-+ AZD-8055

+ Rapamycin

+ DMSO

wt+ DM

SO

TSC2 -/-+ AZD-8055

+ Rapamycin

+ DMSO

***

***

*** ***

*** ***

••

••

Modelling the Neuropathology of Tuberous Sclerosis

( )

Transcrip

tion

alTran

slation

al

f

Growth factorsRapamycin

Control

STAT3

p-STAT3

TSC2 -/-

-+

+- -- -

++

- --

Fig. 4 Inhibition of mTOR corrects translational but not transcriptional dysfunction. a Number of upregulated (gray) or downregulated (black) genes atthe level of translation or transcription in cells treated with rapamycin or AZD-8055 as compared to vehicle-treated cells. b Venn diagrams for genesthat change in gene expression in opposite directions by TSC2 loss (red) and by compound treatments (green, blue), respectively. Overlap highlightshow many genes deregulated in TSC2 depleted cells (upregulated on left, downregulated on right) can be reset to control levels by inhibitor treatment.Thresholds of differentially expressed gene calling: abs(logFC) >= 0.5, counts per million no less than 1, and FDR < 0.05. c Heat map of translationalefficiency for transcripts of ribosomal proteins in control cells and TSC2-deficient cell line (n = 2) after six weeks of differentiation. Vehicle (DMSO)-treated samples reveal the induced TE in TSC2 depleted cell lines that is largely reversed after treatment with either rapamycin or AZD-8055. d Box plotsdisplaying fold changes in transcription, translation, and total protein output for genes associated with proteins synthesis, angiogenesis, and inflamma-tion in control and TSC2-deficient cell lines. While different levels of translation of protein synthesis factors between vehicle-treated control andmutant cells are balanced out by mTOR inhibitor treatment, the different transcriptional and protein output for angiogenesis and inflammationgenes remains unchanged. Values are log2-transformed and normalized to controls. Statistical significance was determined by a two-tailedpaired t-test, ***p < 1×10−6. e Western blot analysis for STAT3 or its phosphorylated form (p-STAT3) from neural cultures after six weeks ofdifferentiation; cultured in the presence (+) or absence (−) of growth factors or rapamycin for the last 12 h before harvest. Elevated phosphorylationeven in the presence of rapamycin indicates continued increased pathway activity. f Molecular mechanisms of the neuropathology of TSC and newtreatment options to be considered

Grabole et al. Genome Medicine (2016) 8:94 Page 11 of 14

Page 12: Genomic analysis of the molecular neuropathology of ...yeolab.github.io/papers/2016/Grabole_GenomeMedicine_2016.pdf · Genomic analysis of the molecular neuropathology of tuberous

differentiation towards a pathologic state. Further, sucha system is well suited to test new treatment strategiesas multiple assays can be performed on material derivedfrom the same source in a reproducible fashion usingisogenic control lines. These advantages may overcomein many instances the inherent limitations of an in vitrosystem. As diverse inter-cellular interactions within thebrain microenvironment are not exactly recreated, notall disease manifestations may be recapitulated. Con-versely, imperfect modeling of physiological conditionscan modify the phenotype making comparison to mousemodels and patient biopsies, as done here, necessary.Overall, this first human stem cell model of TSC willdemonstrate itself to be a valuable system to develop theeffective treatment of cellular hyperplasias in TSC pa-tients based on a better understanding of the diseasebiology.

ConclusionsInsight into the molecular mechanisms underlying theneuropathology of tuberous sclerosis assists a bettermechanistic understanding of the disease and enables de-velopment of novel treatment options for the fullspectrum of clinical manifestations. By investigating ourgenome-edited human neural stem cell model, we wereable to characterize the molecular pathophysiology byprofiling in depth the transcriptional as well as transla-tional changes underlying the disease manifestations.TSC2-deficient cells show the transcriptional signature ofan inflammatory response with implications for epilepto-genesis in TSC patients. In addition, we found intensifiedtranslation of ribosomal proteins, which increases biosyn-thetic capacity in general and boosted production of an-giogenic growth factors specifically. Treatment withmTOR inhibitors reset translational dysfunction but wasincapable to correct transcription-based molecular path-ologies. Importantly, in addition to the detailed descrip-tion of the molecular pathology, we highlight novel entrypoints for tailored pharmacological therapies that hold po-tential to provide substantial benefits to patients sufferingfrom TSC in the future.

Additional files

Additional file 1: Figure S1. Deregulated expression of neuronal andglial markers in the absence of TSC2. (PDF 2.73 mb)

Additional file 2: Figure S2. Loss of TSC2 triggers expression changesrelated to inflammatory response, metabolism, and neuronal function.(PDF 1.15 mb)

Additional file 3: Table S1. RNA-Seq and ribosome profiling data ofdifferentiated TSC2 wild-type, heterozygous, and homozygous deletioncell lines (related to Figs. 2 and 3). (XLSX 6.88 mb)

Additional file 4: Table S2. Gene set enrichment analysis of RNA-Seqdata (related to Additional file 2: Figure S2B and E). (XLSX 22.2 kb)

Additional file 5: Table S3. Gene ontology term enrichments forRNA-Seq data from differentiated TSC2 deletion cell lines and microarraydata of patient cortical tubers (related to Fig. 2e). (XLSX 19.8 kb)

Additional file 6: Table S4. Gene ontology term enrichments forRNA-Seq data from differentiated TSC2 deletion cell lines and microarraydata of patient SEGAs (related to Fig. 2f). (XLSX 27.7 kb)

Additional file 7: Figure S3. Loss of TSC2 increases the TE of factorsimplicated in protein synthesis. (PDF 417 kb)

Additional file 8: Table S5. Gene set enrichment analysis of ribosomeprofiling data (related to Fig. 3d). (XLSX 11.1 kb)

Additional file 9: Table S6. Gene ontology term enrichments forribosome profiling data (related to Fig. 3f). (XLSX 11 kb)

Additional file 10: Table S7. RNA-Seq and ribosome profiling data ofdifferentiated TSC2 wild-type and homozygous deletion cell lines treatedwith mTOR inhibitors (related to Fig. 4). (XLSB 7.52 mb)

Additional file 11: Table S8. Gene sets and associated genes thatshow after mTOR treatment a reversal of the change in expressiondetected in untreated TSC2 deletion cells (related to Fig. 4b). (XLSX 25.8 kb)

AbbreviationsATP: Adenosine triphosphate; BCA: Bicinchoninic assay;cDNA: Complementary DNA; DAPI: 4’,6-diamidino-2-phenylindole;DMSO: Dimethyl sulfoxide; DNA: Desoxyribonuceic acid; ECL: Enhancedchemiluminescence; ESC: Embryonic stem cell; FDR: False discovery rate;GSEA: Gene set enrichment analysis; HRP: Horseradish peroxidase; LOH: Lossof heterozygosity; mTOR: Mechanistic target of rapamycin; NSC: Neural stemcell; OPP: O-propargyl-puromycin; PBS: Phosphate buffered saline;PCR: Polymerase chain reaction; PFA: Paraformaldehyde; qPCR: QuantitativePCR; RNA: Ribonucleic acid; RNA-Seq: RNA-Sequencing; rRNA: RibosomalRNA; SDS-PAGE: Sodium dodecyl sulphate-polyacrylamide gelelectrophoresis; SEGA: Subependymal giant cell astrocytoma; TOP: Terminaloligo-pyrimidine; tRNA: Transfer RNA; TSC: Tuberous sclerosis complex;UTR: Untranslated region

AcknowledgementsNot applicable.

FundingNG, SA, VC, and FW were supported by the Roche Postdoctoral FellowshipProgram. GWY and SA are supported by grants from the National Institutesof Health (HG004659, MH107369, NS075449) and the California Institute ofRegenerative Medicine (RB3-05009 and RB4-06045).

Availability of data and materialsThe datasets generated and analyzed during the current study are availablein the Gene Expression Omnibus repository, http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?token=otstocqydzglxql&acc=GSE78961.

Authors’ contributionsSA, RJ, ME, GWY, and OS conceived the study. NG, SA, and RJ designed theexperiments. NG performed cell differentiation and sequencing experiments.NR and NG performed western blot analysis. VC performedimmunocytochemistry analysis. FCW performed cytokine array analysis. NGand MT performed flow cytometry. NB performed sequencing readprocessing and mapping. JDT performed bioinformatics and statisticalanalysis and data visualization. NG, JDZ, and AK analyzed the data. NG wrotethe manuscript. All authors read and approved the final manuscript.

Competing interestNG, JDZ, NR, VC, FCW, MT, NB, OS, ME, RJ, and AK are employed by F.Hoffmann-La Roche. The remaining authors declare that they have nocompeting interests.

Consent for publicationNot applicable.

Ethics approval and consent to participateNot applicable.

Grabole et al. Genome Medicine (2016) 8:94 Page 12 of 14

Page 13: Genomic analysis of the molecular neuropathology of ...yeolab.github.io/papers/2016/Grabole_GenomeMedicine_2016.pdf · Genomic analysis of the molecular neuropathology of tuberous

Author details1Roche Pharma Research and Early Development, Pharmaceutical Sciences,Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse124, Basel 4070, Switzerland. 2Department of Cellular and MolecularMedicine, Institute for Genomic Medicine, UCSD Stem Cell Program, SanfordConsortium for Regenerative Medicine, University of California at San Diego,La Jolla, CA 92037, USA. 3Roche Pharma Research and Early Development,Neuroscience, Ophthalmology and Rare Diseases, Roche Innovation CenterBasel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070,Switzerland.

Received: 20 April 2016 Accepted: 18 August 2016

References1. Laplante M, Sabatini DM. mTOR signaling in growth control and disease.

Cell. 2012;149:274–93.2. Crino PB, Nathanson KL, Henske EP. The tuberous sclerosis complex. N Engl

J Med. 2006;355:1345–56.3. Curatolo P, Bombardieri R, Jozwiak S. Tuberous sclerosis. Lancet. 2008;372:

657–68.4. Rosser T, Panigrahy A, McClintock W. The diverse clinical manifestations of

tuberous sclerosis complex: a review. Semin Pediatr Neurol. 2006;13:27–36.5. Chan JA, Zhang H, Roberts PS, Jozwiak S, Wieslawa G, Lewin-Kowalik J,

et al. Pathogenesis of tuberous sclerosis subependymal giant cellastrocytomas: biallelic inactivation of TSC1 or TSC2 leads to mTORactivation. J Neuropathol Exp Neurol. 2004;63:1236–42.

6. Henske EP, Wessner LL, Golden J, Scheithauer BW, Vortmeyer AO, Zhuang Z,et al. Loss of tuberin in both subependymal giant cell astrocytomas andangiomyolipomas supports a two-hit model for the pathogenesis oftuberous sclerosis tumors. Am J Pathol. 1997;151:1639–47.

7. Crino PB, Aronica E, Baltuch G, Nathanson KL. Biallelic TSC gene inactivationin tuberous sclerosis complex. Neurology. 2010;74:1716–23.

8. DiMario FJ. Brain abnormalities in tuberous sclerosis complex. J ChildNeurol. 2004;19:650–7.

9. Franz DN, Leonard J, Tudor C, Chuck G, Care M, Sethuraman G, et al.Rapamycin causes regression of astrocytomas in tuberous sclerosis complex.Ann Neurol. 2006;59:490–8.

10. Franz DN, Belousova E, Sparagana S, Bebin EM, Frost M, Kuperman R, et al.Efficacy and safety of everolimus for subependymal giant cell astrocytomasassociated with tuberous sclerosis complex (EXIST-1): a multicentre,randomised, placebo-controlled phase 3 trial. Lancet. 2013;381:125–32.

11. Zhou J, Shrikhande G, Xu J, McKay RM, Burns DK, Johnson JE, et al.Tsc1 mutant neural stem/progenitor cells exhibit migration deficits andgive rise to subependymal lesions in the lateral ventricle. Genes Dev.2011;25:1595–600.

12. Magri L, Cambiaghi M, Cominelli M, Alfaro-Cervello C, Cursi M, Pala M, et al.Sustained activation of mTOR pathway in embryonic neural stem cells leadsto development of tuberous sclerosis complex-associated lesions. Cell StemCell. 2011;9:447–62.

13. Carson RP, Van Nielen DL, Winzenburger PA, Ess KC. Neuronal and gliaabnormalities in Tsc1-deficient forebrain and partial rescue by rapamycin.Neurobiol Dis. 2012;45:369–80.

14. Ess KC, Uhlmann EJ, Li W, Li H, Declue JE, Crino PB, et al. Expressionprofiling in tuberous sclerosis complex (TSC) knockout mouse astrocytes tocharacterize human TSC brain pathology. Glia. 2004;46:28–40.

15. Onda H. Tsc2 null murine neuroepithelial cells are a model for human tubergiant cells, and show activation of an mTOR pathway. Mol Cell Neurosci.2002;21:561–74.

16. Lee D-F, Su J, Kim HS, Chang B, Papatsenko D, Zhao R, et al. Modelingfamilial cancer with induced pluripotent stem cells. Cell. 2015;161:240–54.

17. Sosunov AA, Wu X, Weiner HL, Mikell CB, Goodman RR, Crino PD, et al.Tuberous sclerosis: a primary pathology of astrocytes? Epilepsia. 2008;49Suppl 2:53–62.

18. Costa V, Aigner S, Vukcevic M, Sauter E, Behr K, Ebeling M, et al. mTORC1inhibition corrects neurodevelopmental and synaptic alterations in a humanstem cell model of tuberous sclerosis. Cell Rep. 2016;15:86–95.

19. Trapnell C, Pachter L, Salzberg SL. TopHat: discovering splice junctions withRNA-Seq. Bioinformatics. 2009;25:1105–11.

20. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, et al. The SequenceAlignment/Map format and SAMtools. Bioinformatics. 2009;25:2078–9.

21. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package fordifferential expression analysis of digital gene expression data.Bioinformatics. 2009;26:139–40.

22. Wu D, Smyth GK. Camera: a competitive gene set test accounting for inter-gene correlation. Nucleic Acids Res. 2012;40:e133–3.

23. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powersdifferential expression analyses for RNA-sequencing and microarray studies.Nucleic Acids Res. 2015;43:e47–7.

24. Olshen AB, Hsieh AC, Stumpf CR, Olshen RA, Ruggero D, Taylor BS.Assessing gene-level translational control from ribosome profiling.Bioinformatics. 2013;29:2995–3002.

25. Yuan SH, Martin J, Elia J, Flippin J, Paramban RI, Hefferan MP, et al.Cell-surface marker signatures for the isolation of neural stem cells, gliaand neurons derived from human pluripotent stem cells. PLoS ONE.2011;6:e17540–16.

26. Hockemeyer D, Soldner F, Beard C, Gao Q, Mitalipova M, DeKelver RC, et al.Efficient targeting of expressed and silent genes in human ESCs and iPSCsusing zinc-finger nucleases. Nat Biotechnol. 2009;27:851–7.

27. Dunkley T, Costa V, Friedlein A, Lugert S, Aigner S, Ebeling M, et al.Characterization of a human pluripotent stem cell-derived model ofneuronal development using multiplexed targeted proteomics. ProteomicsClin Appl. 2015;9:684–94.

28. Way SW, McKenna J, Mietzsch U, Reith RM, Wu HCJ, Gambello MJ. Loss ofTsc2 in radial glia models the brain pathology of tuberous sclerosis complexin the mouse. Hum Mol Genet. 2009;18:1252–65.

29. Mietzsch U, McKenna III J, Reith RM, Way SW, Gambello MJ. Comparativeanalysis of Tsc1and Tsc2single and double radial glial cell mutants. J CompNeurol. 2013;521:3817–31.

30. Meikle L, Talos DM, Onda H, Pollizzi K, Rotenberg A, Sahin M, et al. A mousemodel of tuberous sclerosis: neuronal loss of TSC1 causes dysplastic andectopic neurons, reduced myelination, seizure activity, and limited survival.J Neurosci. 2007;27:5546–58.

31. Feliciano DM, Su T, Lopez J, Platel J-C, Bordey A. Single-cell Tsc1 knockoutduring corticogenesis generates tuber-like lesions and reduces seizurethreshold in mice. J Clin Invest. 2011;121:1596–607.

32. Boer K, Troost D, Jansen F, Nellist M, van den Ouweland AMW, Geurts JJG,et al. Clinicopathological and immunohistochemical findings in an autopsycase of tuberous sclerosis complex. Neuropathology. 2008;28:577–90.

33. He F, Ge W, Martinowich K, Becker-Catania S, Coskun V, Zhu W, et al.A positive autoregulatory loop of Jak-STAT signaling controls the onset ofastrogliogenesis. Nat Neurosci. 2005;8:616–25.

34. Herrmann JE, Imura T, Song B, Qi J, Ao Y, Nguyen TK, et al. STAT3 is acritical regulator of astrogliosis and scar formation after spinal cord injury.J Neurosci. 2008;28:7231–43.

35. Hill MO. Correspondence analysis: a neglected multivariate method. ApplStat. 1974;23:340–54.

36. Zamanian JL, Xu L, Foo LC, Nouri N, Zhou L, Giffard RG, et al. Genomicanalysis of reactive astrogliosis. J Neurosci. 2012;32:6391–410.

37. Boer K, Crino PB, Gorter JA, Nellist M, Jansen FE, Spliet WGM, et al. Geneexpression analysis of tuberous sclerosis complex cortical tubers revealsincreased expression of adhesion and inflammatory factors. Brain Pathol.2010;20:704–19.

38. Boer K, Jansen F, Nellist M, Redeker S, van den Ouweland AMW, SplietWGM, et al. Inflammatory processes in cortical tubers and subependymalgiant cell tumors of tuberous sclerosis complex. Epilepsy Res. 2008;78:7–21.

39. Maldonado M, Baybis M, Newman D, Kolson DL, Chen W, McKhann II G,et al. Expression of ICAM-1, TNF-α, NFkB, and MAP kinase in tubers of thetuberous sclerosis complex. Neurobiol Dis. 2003;14:279–90.

40. Iyer AM, Zurolo E, Boer K, Baayen JC, Giangaspero F, Arcella A, et al. Tissueplasminogen activator and urokinase plasminogen activator in humanepileptogenic pathologies. NSC. 2010;167:929–45.

41. Shu H-F, Zhang C-Q, Yin Q, An N, Liu S-Y, Yang H. Expression of theinterleukin 6 system in cortical lesions from patients with tuberous sclerosiscomplex and focal cortical dysplasia type IIb. J Neuropathol Exp Neurol.2010;69:838–49.

42. Lesma E, Ancona S, Sirchia SM, Orpianesi E, Grande V, Colapietro P, et al.TSC2epigenetic defect in primary LAM cells. Evidence of an anchorage-independent survival. J Cell Mol Med. 2014;18:766–79.

43. Zhang B, Zou J, Rensing NR, Yang M, Wong M. Inflammatory mechanismscontribute to the neurological manifestations of tuberous sclerosis complex.Neurobiol Dis. 2015;80:70–9.

Grabole et al. Genome Medicine (2016) 8:94 Page 13 of 14

Page 14: Genomic analysis of the molecular neuropathology of ...yeolab.github.io/papers/2016/Grabole_GenomeMedicine_2016.pdf · Genomic analysis of the molecular neuropathology of tuberous

44. Gingras AC, Raught B, Sonenberg N. Regulation of translation initiation byFRAP/mTOR. Genes Dev. 2001;15:807–26.

45. Ingolia NT, Lareau LF, Weissman JS. Ribosome profiling of mouse embryonicstem cells reveals the complexity and dynamics of mammalian proteomes.Cell. 2011;147:789–802.

46. Thoreen CC, Chantranupong L, Keys HR, Wang T, Gray NS, Sabatini DM.A unifying model for mTORC1-mediated regulation of mRNA translation.Nature. 2012;486:109–13.

47. Hsieh AC, Liu Y, Edlind MP, Ingolia NT, Janes MR, Sher A, et al. Thetranslational landscape of mTOR signalling steers cancer initiation andmetastasis. Nature. 2012;485:55–61.

48. Truitt ML, Conn CS, Shi Z, Pang X, Tokuyasu T, Coady AM, et al. Differentialrequirements for eIF4E dose in normal development and cancer. Cell. 2015;162:59–71.

49. Ruggero D, Pandolfi PP. Does the ribosome translate cancer? Nat RevCancer. 2003;3:179–92.

50. Ruggero D. Translational control in cancer etiology. Cold Spring HarbPerspect Biol. 2013;5:a012336.

51. Arbiser JL, Brat D, Hunter S, D’Armiento J, Henske EP, Arbiser ZK, et al.Tuberous sclerosis-associated lesions of the kidney, brain, and skin areangiogenic neoplasms. J Am Acad Dermatol. 2002;46:376–80.

52. Sun S, Chen S, Liu F, Wu H, McHugh J, Bergin IL, et al. Constitutiveactivation of mTORC1 in endothelial cells leads to the development andprogression of lymphangiosarcoma through VEGF autocrine signaling.Cancer Cell. 2015;28:758–72.

53. Krueger DA, Wilfong AA, Holland-Bouley K, Anderson AE, Agricola K, TudorC, et al. Everolimus treatment of refractory epilepsy in tuberous sclerosiscomplex. Ann Neurol. 2013;74:679–87.

54. Chresta CM, Davies BR, Hickson I, Harding T, Cosulich S, Critchlow SE, et al.AZD8055 is a potent, selective, and orally bioavailable ATP-competitivemammalian target of rapamycin kinase inhibitor with in vitro and in vivoantitumor activity. Cancer Res. 2010;70:288–98.

55. Pekny M, Nilsson M. Astrocyte activation and reactive gliosis. Glia. 2005;50:427–34.56. Krencik R, Ullian EM. A cellular star atlas: using astrocytes from human

pluripotent stem cells for disease studies. Front Cell Neurosci. 2013;7:25.57. Sloan SA, Barres BA. Mechanisms of astrocyte development and their contributions

to neurodevelopmental disorders. Curr Opin Neurobiol. 2014;27:75–81.58. Hegedus B, Dasgupta B, Shin JE, Emnett RJ, Hart-Mahon EK, Elghazi L, et al.

Neurofibromatosis-1 regulates neuronal and glial cell differentiation fromneuroglial progenitors in vivo by both cAMP- and Ras-dependentmechanisms. Cell Stem Cell. 2007;1:443–57.

59. Paquin A, Hordo C, Kaplan DR, Miller FD. Costello syndrome H-Ras allelesregulate cortical development. Dev Biol. 2009;330:440–51.

60. Gauthier AS, Furstoss O, Araki T, Chan R, Neel BG, Kaplan DR, et al. Controlof CNS cell-fate decisions by SHP-2 and its dysregulation in Noonansyndrome. Neuron. 2007;54:245–62.

61. Urosevic J, Sauzeau V, Soto-Montenegro ML, Reig S, Desco M, Wright EMB,et al. Constitutive activation of B-Raf in the mouse germ line provides a modelfor human cardio-facio-cutaneous syndrome. Proc Natl Acad Sci U S A. 2011;108:5015–20.

• We accept pre-submission inquiries

• Our selector tool helps you to find the most relevant journal

• We provide round the clock customer support

• Convenient online submission

• Thorough peer review

• Inclusion in PubMed and all major indexing services

• Maximum visibility for your research

Submit your manuscript atwww.biomedcentral.com/submit

Submit your next manuscript to BioMed Central and we will help you at every step:

Grabole et al. Genome Medicine (2016) 8:94 Page 14 of 14