<<

Genome editing and for disease modeling Adriana S. Beltran Assistant Professor, Department of Pharmacology Director, Human Pluripotent Stem Cell Core Phone: (919) 537-3996 (office), (919) 537-3995 (lab) Email: [email protected] Website: http://www.med.unc.edu/humanstemcellcore The University of North Carolina at Chapel Hill

April 13, 2017 The human Pluripotent Stem Cell core (HPSCC) is committed to accelerating collaborative studies in stem

The HPSCC mission is to serve as the central platform of technical support for investigators seeking to work with human embryonic and pluripotent stem cells Pluripotent stem cells

Embryonic Stem cells

Self-renew Differen5a5on

PROS Limitless poten5al in regenera5ve medicine and disease

CONS Rejec5on aBer transplanta5on Usage of human embryos to create them Inducing a pluripotent state (iPS)

Self-renewal network-ON Self-renewal network-OFF Differentiation genes-OFF Differentiation genes-ON

ES Cell Differentiation

Somatic cells iPS Cell Reprogramming

Oct4 Klf4 Sox2 c-Myc

464

has ASD technology editing Gene . [22,33] ) 3 Box ( possible become associated are that measures Quantifiable iPSCs.

have performed iPSCs within genes single manipulating improved, by manifested phenotypes vitro in examine to

or lowing (TALEN) nucleases effector activator-like transcription be can analysis functional differentiation, neuronal

editing by (ZFN) nuclease finger zinc as such technologies Fol- patients. ASD from cells somatic reprogramming

modeling murine gene As . [32] iPSCs using disorders X-linked available readily are iPSCs Meanwhile, . [9] models

inactivation gical for concern a raising culture, in time over transgenic by examined be can genes the of functions

X can of erosion undergoes iPSCs female in chromosome physiolo- the and genes candidate culprit the identify

. [28–31] When inactive the that showed study recent a However, methods genomics ASD, with diagnosed is person a

) 2 Box and below (see feature this of advantage taking iPSCs. and models, animal genomics, using ASD studying

by patients syndrome Rett female from chromosome X for scheme a depicts 1 Figure . [26] cells heterologous of use

active the on allele mutant or type wild the either with the prevent that barriers immunological and ESCs human

produced be can iPSCs Thus, . [27] fibroblasts of status obtain to embryos destroying with associated issues ethical

chromosome X active/repressed the retaining by iPSCs the bypass iPSCs Furthermore, therapies. replacement

female isogenic of sets unique produces reprogramming cell for sources autologous and screening, drug for forms

females, in prominent and X-linked is disease a when that plat- models, cellular disease-specific of development the

fact the of advantage take to is lines isogenic generating enable patients from iPSCs Thus, . [20–25] cells parental

for method One control. ideal the are lines iPSC isogenic by manifested those represent presumably iPSCs from

Nevertheless, effect. genetic compounding the reduce differentiated cells of phenotypes the cells, somatic ental

can parents, or siblings as such persons, related genetically par- the of composition genetic the retain iPSCs Because

closely from derived iPSCs of use the physiology, cellular self-renewal. for capacity the and potential, differentiation

individual morphology, on influence large a have variations genetic pluripotent markers, surface of expression

healthy Klf-4 Because . [21] controls as used been have persons as such characteristics share ESCs and iPSCs . [18,19]

from iPSCs often, Most phenotypes. vitro in the of pretation and , c-Myc , Sox2 , Oct4 factors transcription four the of sion

inter- the determines control standard of selection proper expres- ectopic the by cells differentiated reprogramming

the ASD, for model cellular a as iPSCs using When by created been have iPSCs . [14–17] tissues and organs

ASD. human damaged treating as well as diseases

human 464

ameliorating for chemicals of efficacy the test to platform modeling for resources cellular considered are ESCs

has ASD technology editing Gene . [22,33] ) 3 Box ( possible become associated are that measures Quantifiable

iPSCs.

screening a as used be can migration, neuronal and ity, capacities, self-renewing and pluripotent their to Owing ).

1

have performed iPSCs within genes single manipulating improved, by manifested phenotypes vitro in examine

to

activ- synaptic connectivity, neuronal as such ASDs, with Box ( layers germ three all to rise give can they

pluripotent

or lowing (TALEN) nucleases effector activator-like transcription be can analysis functional differentiation, neuronal

editing by (ZFN) nuclease finger zinc as such technologies Fol- patients. ASD from cells somatic reprogramming

modeling murine gene As . [32] iPSCs using disorders X-linked available readily are iPSCs Meanwhile, . [9] models

inactivation gical for concern a raising culture, in time over transgenic by examined be can genes the of functions

therapy. cell-based or modeling disease vitro in for controls

X can of erosion undergoes iPSCs female in chromosome physiolo- the and genes candidate culprit the identify

isogenic provide to given the correcting facilitate technologies editing gene developed Recently treatments. potential as drugs screen to and pathogenesis

. [28–31] When inactive the that showed study recent a However, methods genomics ASD, with diagnosed is person a

disease novel elucidate to used be can cells neuronal The ). 1 Table ( protocols well-established using iPSCs from directed be can ASD, for relevant

iPSC as patient-specific pre-clinical most

) 2 Box and below (see feature this of advantage taking iPSCs. and models, animal genomics, using ASD

studying

are which neurons, Cortical generated. be can iPSCs ASD patient-derived samples, blood or biopsy skin From genes. ASD novel seek actively to used are tools

genomics

by patients syndrome Rett female from chromosome X for scheme a depicts 1 Figure . [26] cells heterologous of

use

Advanced defects. genetic well-defined by caused are syndrome, Timothy or syndrome, X Fragile syndrome, Rett including ASDs, syndromic Monogenic

disorder.

active the on allele mutant or type wild the either with the prevent that barriers immunological and ESCs

human neurodevelopmental genetic a is ASD research. (ASD) disorder spectrum autism in technology (iPSC) cell stem pluripotent induced adopting of Overview . 1

Figure

produced be can iPSCs Thus, . [27] fibroblasts of status obtain to embryos destroying with associated issues

disease models ethical

TRENDS in Molecular Medicine Medicine Molecular in TRENDS

chromosome X active/repressed the retaining by iPSCs the bypass iPSCs Furthermore, therapies. Review Trends in Molecular Medicine August 2012, Vol. 18, No. 8 replacement

female isogenic of sets unique produces reprogramming cell for sources autologous and screening, drug for forms

females, in prominent and X-linked is disease a when that plat- models, cellular disease-specific of development

the

model

fact the of advantage take to is lines isogenic generating enable patients from iPSCs Thus, . [20–25] cells

parental

Animal

for method One control. ideal the are lines iPSC isogenic by manifested those represent presumably iPSCs from

Nevertheless, effect. genetic compounding the reduce differentiated cells of phenotypes the cells, somatic ental Timothy syndrome, etc. syndrome, Timothy

can parents, or siblings as such persons, related genetically par- the of composition genetic the retain iPSCs

Because

gene causing ASD causing gene

Fragile X syndrome X Fragile

Review Trends in Molecular Medicine August 2012, Vol. 18, No. 8

closely from derived iPSCs of use the physiology, cellular self-renewal. for capacity the and potential, differentiation on of novel of on  fica  Iden syndrome,  Re

individual morphology, on influence large a have variations genetic pluripotent markers, surface of

expression Monogenic disorders: Monogenic

Gene engineering Gene

healthy Klf-4 Because . [21] controls as used been have persons as such characteristics share ESCs and iPSCs . [18,19]

from iPSCs often, Most phenotypes. vitro in the of pretation and , c-Myc , Sox2 , Oct4 factors transcription four the of sion

inter- the determines control standard of selection proper expres- ectopic the by cells differentiated

reprogramming

and genomics Molecular genomics and therapy Disease the ASD, for model cellular a as iPSCs using When by created been have iPSCs . [14–17] tissues and Somac cells organs

Disease cs  gene in Advances

Cell ASD. human damaged treating as well as diseases modeling mechanism of human

modeling ameliorating for chemicals of efficacy the test to platform modeling for resources cellular considered are

Specific Neuronal ESCs

the disease

ent ent  pa ASD

Reprograming iPSC Disease screening a as used be can migration, neuronal and ity, capacities, self-renewing and pluripotent their to Owing ). Somac cells 1

differen9a9on

differenaonmodeling

activ- synaptic connectivity, neuronal as such ASDs, with Box ( layers germ three all to rise give can they Reprogramming pluripotent

discovery Neuronal

Drug differenaon

Reprogramming Drug

Reprogramming Reprogramming

on on  a  eren ff

di Drugdiscovery

Neuronal therapy. cell-based or modeling disease vitro in for discovery controls

modeling isogenic provide to mutation given the correcting facilitate technologies editing gene developed Recently treatments. potential as drugs screen to and iPSCs pathogenesis

iPSC

disease novel elucidate to used be can cells neuronal The ). 1 Table ( protocols well-established using iPSCs from directed be can ASD, for neurons relevant most c cells cells c  ASD paent Soma

iPSC Specific cell type ASD paent Disease

are which neurons, Cortical generated. be can iPSCs ASD patient-derived samples, blood or biopsy skin From genes. ASD novel seek actively to used are tools

differen9a9on genomics Advanced defects. genetic well-defined by caused are syndrome, Timothy or syndrome, X Fragile syndrome, Rett including ASDs, syndromic Monogenic

Advances in genecs Cell Cell Cardiac, neural, disorder. neurodevelopmental genetic a is ASD research. (ASD) disorder spectrum autism in technology (iPSC) cell stem pluripotent induced adopting of Overview . 1 Drug Figure Advances in genecs therapy

and genomics therapydiscovery liver toxicity and genomics Medicine Molecular in TRENDS tests

Gene engineering

Pa5ents Monogenic disorders: model Gene9c engineering Re syndrome, Idenficaon of novel Animal

Fragile X syndrome gene causing ASD Gene engineering

Monogenic disorders: etc. syndrome, Timothy

Timothy syndrome, etc. 8 No. 18, Vol. 2012, August Medicine Molecular in Trends gene causing ASD causing gene Fragile X syndrome X Fragile

Re syndrome, Idenficaon of novel Review

on of novel of on  fica  Iden

syndrome, 

Animal Re

Monogenic disorders: Monogenic Fragile X syndrome gene causing ASD engineering Gene model

Timothy syndrome, etc.

and genomics genomics and

therapy TRENDS in Molecular Medicine Regenera9ve cs  gene in Advances

Cell Animal

Figure 1. Overview of adopting induced pluripotent stem cell (iPSC) technology in autism spectrum disorder (ASD) research. ASD is a genetic neurodevelopmentalMedicine

ent ent  model pa ASD

iPSC

disorder. Monogenic syndromic ASDs, including Rett syndrome, Fragile X syndrome, or Timothy syndrome, areSpecific cell type caused by well-defined genetic defects. Advanced

genomics tools are used to actively seek novel ASD genes. From skin biopsy or blood samples, patient-derived ASD iPSCs can be generated. Cortical neurons, which are

discovery differen9a9on

most relevant neurons for ASD, can be directed from iPSCs using well-established protocols (Table 1). The neuronal cells can be used to elucidate novel disease

Drug

pathogenesis and to screen drugs as potential treatments. Recently developed gene editing technologies facilitate correcting the given mutation to provide isogenic

Reprogramming controls for in vitro disease modeling or cell-based therapy. TRENDS in Molecular Medicine on on  a  eren ff di

Neuronal

modeling

Figure 1. Overview of adopting induced pluripotent stem cell (iPSC) technology in autism spectrum disorder (ASD) research. ASD is a genetic neurodevelopmental c cells cells c  Soma

Disease

disorder. Monogenic syndromic ASDs, including Rett syndrome, Fragile X syndrome, or Timothy syndrome, are caused by well-defined genetic defects. Advanced

pluripotent they can give rise to all three germ layers (Box with ASDs, such as neuronal connectivity, synaptic activ-

genomics tools are used to actively seek novel ASD genes. From skin biopsy or blood samples, patient-derived ASD iPSCs can be generated. Cortical neurons, which are

1). Owing to their pluripotent and self-renewing capacities, ity, and neuronal migration, can be used as a screening

most relevant neurons for ASD, can be directed from iPSCs using well-established protocols (Table 1). The neuronal cells can be used to elucidate novel disease

ESCs are considered cellular resources for modeling platform to test the efficacy of chemicals for ameliorating

pathogenesis and to screen drugs as potential treatments. Recently developed gene editing technologies facilitate correcting the given mutation to provide isogenic

human diseases as well as treating damaged human ASD.

controls for in vitro disease modeling or cell-based therapy.

organs and tissues [14–17]. iPSCs have been created by When using iPSCs as a cellular model for ASD, the

reprogramming differentiated cells by the ectopic expres- proper selection of standard control determines the inter- 8 No. 18, Vol. 2012, August Medicine Molecular in Trends

Review

sion of the four transcription factors Oct4, Sox2, c-Myc, and pretation of the in vitro phenotypes. Most often, iPSCs from

Klf-4 [18,19]. iPSCs and ESCs share characteristics such as healthy persons have been used as controls [21]. Because

morphology, expression of surface markers, pluripotent individual genetic variations have a large influence on

pluripotent theydifferentiationcan give rise potential,to all threeand thegermcapacitylayersfor self-renewal.(Box withcellularASDs,physiology,such astheneuronaluse of iPSCsconnectivity,derived fromsynapticclosely activ-

1). Owing to theirBecausepluripotentiPSCs retainand self-renewingthe genetic compositioncapacities,of the par-ity,geneticallyand neuronalrelated persons,migration,such ascansiblingsbe usedor parents,as acanscreening

ental somatic cells, the phenotypes of cells differentiated reduce the compounding genetic effect. Nevertheless,

ESCs are considered cellular resources for modeling platform to test the efficacy of chemicals for ameliorating

from iPSCs presumably represent those manifested by isogenic iPSC lines are the ideal control. One method for

human diseasesparentalas wellcells [20–25]as treating. Thus, iPSCsdamagedfrom patientshumanenableASD.generating isogenic lines is to take advantage of the fact

the development of disease-specific cellular models, plat- that when a disease is X-linked and prominent in females,

organs and tissues [14–17]. iPSCs have been created by When using iPSCs as a cellular model for ASD, the

forms for drug screening, and autologous sources for cell reprogramming produces unique sets of isogenic female

reprogrammingreplacementdifferentiated therapies.cellsFurthermore,by the ectopiciPSCsexpres-bypass theproperiPSCsselectionby retainingof standardthe active/repressedcontrol determinesX chromosomethe inter-

ethical issues associated with destroying embryos to obtain status of fibroblasts [27]. Thus, iPSCs can be produced

sion of the four transcription factors Oct4, Sox2, c-Myc, and pretation of the in vitro phenotypes. Most often, iPSCs from

human ESCs and immunological barriers that prevent the with either the wild type or mutant allele on the active

Klf-4 [18,19]. iPSCsuse ofandheterologousESCs sharecells [26]characteristics. Figure 1 depictssucha schemeas forhealthyX chromosomepersons fromhavefemalebeen Rettusedsyndromeas controlspatients[21]by. Because

morphology, expressionstudying ASDofusingsurfacegenomics,markers,animal models,pluripotentand iPSCs.individualtaking advantagegeneticofvariationsthis feature have(see belowa largeand Boxinfluence2) on

When a person is diagnosed with ASD, genomics methods [28–31]. However, a recent study showed that the inactive

differentiation potential, and the capacity for self-renewal. cellular physiology, the use of iPSCs derived from closely

can identify the culprit candidate genes and the physiolo- X chromosome in female iPSCs undergoes erosion of

Because iPSCs gicalretainfunctionsthe geneticof the genescompositioncan be examinedof theby transgenicpar- geneticallyinactivationrelatedover timepersons,in culture,suchraisingas siblingsa concernor parents,for can

murine models [9]. Meanwhile, iPSCs are readily available modeling X-linked disorders using iPSCs [32]. As gene

ental somatic cells, the phenotypes of cells differentiated reduce the compounding genetic effect. Nevertheless,

by reprogramming somatic cells from ASD patients. Fol- editing technologies such as zinc finger nuclease (ZFN)

from iPSCs presumablylowing neuronalrepresentdifferentiation,thosefunctionalmanifestedanalysisbycan beisogenicor transcriptioniPSC linesactivator-likeare the effectorideal control.nucleases One(TALEN)method for

performed to examine in vitro phenotypes manifested by have improved, manipulating single genes within iPSCs

parental cells [20–25]. Thus, iPSCs from patients enable generating isogenic lines is to take advantage of the fact

ASD iPSCs. Quantifiable measures that are associated has become possible (Box 3) [22,33]. Gene editing technology

the development of disease-specific cellular models, plat- that when a disease is X-linked and prominent in females,

464

forms for drug screening, and autologous sources for cell reprogramming produces unique sets of isogenic female

replacement therapies. Furthermore, iPSCs bypass the iPSCs by retaining the active/repressed X chromosome

ethical issues associated with destroying embryos to obtain status of fibroblasts [27]. Thus, iPSCs can be produced

human ESCs and immunological barriers that prevent the with either the wild type or mutant allele on the active

use of heterologous cells [26]. Figure 1 depicts a scheme for X chromosome from female Rett syndrome patients by

studying ASD using genomics, animal models, and iPSCs. taking advantage of this feature (see below and Box 2)

When a person is diagnosed with ASD, genomics methods [28–31]. However, a recent study showed that the inactive

can identify the culprit candidate genes and the physiolo- X chromosome in female iPSCs undergoes erosion of

gical functions of the genes can be examined by transgenic inactivation over time in culture, raising a concern for

murine models [9]. Meanwhile, iPSCs are readily available modeling X-linked disorders using iPSCs [32]. As gene

by reprogramming somatic cells from ASD patients. Fol- editing technologies such as zinc finger nuclease (ZFN)

lowing neuronal differentiation, functional analysis can be or transcription activator-like effector nucleases (TALEN)

performed to examine in vitro phenotypes manifested by have improved, manipulating single genes within iPSCs

ASD iPSCs. Quantifiable measures that are associated has become possible (Box 3) [22,33]. Gene editing technology

464 Genome editing in rats 47

cussed in relation to genetic analysis and manipulation in KI . Therefore, artificially designed ZFNs in animals, especially in rats. can be used to generate KO or KI alleles at the tar- geted sequences via NHEJ or HR repair, respectively. A summary of how to generate targeted KO rats Zinc-finger nucleases using ZFNs is given in Figure. 1. Briefly, two ZFNs are Zinc finger nucleases are chimeric proteins that consist designed across the spacer domain to recognize the of a specific DNA-binding domain that is made of tan- targeted DNA sequences. Messenger RNAs are then dem zinc finger-binding motifs fused to a non-specific transcribed in vitro from the two ZFN plasmids and cleavage domain of the restriction endonuclease FokI injected into the male pronuclei of rat . Pronu- (Bibikova et al. 2001; Porteus & Carroll 2005; Wu et al. clear stage embryos are collected from female rats 2007) (Fig. 1). As one zinc finger unit binds with 3-bp that were superovulated by equine chorionic gonado- of DNA, 9–18 bp sequences can be specifically recog- tropin and human chorionic gonadotropin injection. nized by combining 3–6 different zinc finger units. By The ZFN-injected embryos that differentiate into two designing two zinc finger motifs on either side of 5– cells are then transferred to the oviduct of pseudo- 6 bp spacer sequences at a target region, the FokI pregnant females. This method is based on a similar nuclease combined with the zinc finger can introduce technique used to produce conventional transgenic a double-strand break (DSB) within the 5–6 bp spacer animals (Palmiter et al. 1982, 1983; Mullins et al. sequences. Although the DSB is usually repaired via 1990), except that mRNA is used. The procedure for non-homologous end joining (NHEJ), an arbitrary dele- the micromanipulation of embryos is the same for all tion or deletion of base pairs often occurs during the nucleases, including the below-mentioned TALEN and repair process. Consequently, repair by NHEJ is muta- clustered regularly interspaced short palindromic genic and mostly results in a loss-of-function mutation. repeats (CRISPR) enzymes. Moreover, if DNA fragments homologous to the tar- The ZFN technology was first reported in the 1990s geted sequences are co-injected with the nucleases, (Kim et al. 1996; Chandrasegaran & Smith 1999). homologous recombination (HR) can occur, enabling From the 2000s, ZFNs have been developed for insertionTools of a transgene orfor replacement genome of the homolo- various mammalian editing cells (Bibikova et al. 2001, 2003; gous sequences at the targeted region, which results Porteus & Baltimore 2003; Urnov et al. 2005;

Zinc Finger nucleases (ZFN)

FokI nuclease FOX1

GCCAACCGCACTATGCCTCTATAACTACCGGAGCAGTCGAGGTTGAGCTT CGGTTGGCGTGATACGGAGATATTGATGGCCTCGTCAGCTCCAACTCGAA

FOX1 FokI nuclease

Easy to design Advantage: Long experience in using Simplicity Clinical trial High specificity High efficiency

Labor intense to construct Disadvantage: Difficult to designed and PAM requirement “T” requirement Construct Off-target concerns Target site restric9ons Sensi9ve to 5’methylcytosine

Fig. 1. Gene targeting technologies with zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clus- tered regularly interspaced short palindromic repeats (CRISPR)/Cas in rats. Schematic representation of the methods used for generating targeted knockout rats.

Mashimo T. 2014, 56, 46-52 ª 2013 The Author Development, Growth & Differentiation ª 2013 Japanese Society of Developmental Biologists Medical applications of iPS cells REVIEW INSIGHT Figure 2 | Medical applications of iPS cells. Reprogramming technology and iPS cells have the potential to be used to model and treat human disease. In this example, the patient has Treatment Transplantation of genetically a neurodegenerative disorder. Patient-specific with drugs matched healthy cells iPS cells — in this case derived by ectopic co-expression of transcription factors in cells isolated from a skin biopsy — can be used in one of two pathways. In cases in which the disease- Patient causing mutation is known (for example, familial Parkinson’s disease), gene targeting could be Disease-specifc drugs used to repair the DNA sequence (right). The cMYC OCT4 Healthy cells gene-corrected patient-specific iPS cells would then undergo directed differentiation into the Screeningening fforor KLF4 SOX2 affected neuronal subtype (for example, midbrain therapeuticapeutic In vitro compoundspounds diferentiation dopaminergic neurons) and be transplanted into the patient’s brain (to engraft the nigrostriatal axis). Alternatively, directed differentiation of the patient-specific iPS cells into the affected Skin biopsy, blood, kera5nocytes Skin biopsy neuronal subtype (left) will allow the patient’s disease to be modelled in vitro, and potential drugs Afected cell type Repaired iPS cells can be screened, aiding in the discovery of novel therapeutic compounds.

Use gene targeting to repair In vitro disease-causing mutation diferentiation

Patient-specifc iPS cells

Given the number of drugs that have notoriously been withdrawn from this study, we showed that a high expression level of multiple telomerase the market because of their tendency to induce arrhythmias, it is highly components was characteristic of the pluripotent state more generally, Adapted from: Robinton & Daley, Nature 2012 likely that the current inadequate approaches for assessing cardiotoxic- illustrating how iPS cells can reveal fundamental aspects of cell biology. ity will be complemented by iPS-cell-based assessments of drug effects. An independent study of the reprogramming of cells from patients with A study from our laboratory explored dyskeratosis congenita, a dis- dyskeratosis congenita confirmed the general transcriptional upregula- order of telomere maintenance, and provided an unanticipated insight tion of multiple telomerase components and the maintenance of telomere into the basic biology of telomerase that has therapeutic implications73. lengths in clones74; however, in this study, no clones with elongated telom- In its most severe form, dyskeratosis congenita is caused by a mutation eres were identified. The different outcomes of these studies highlight the in the dyskerin gene (DKC1), which is X linked, leading to shortened tel- limitations of iPS-cell-based disease models that are imposed by clonal omeres and premature senescence in cells and ultimately manifesting as variation as a result of the inherent technical infidelity of reprogram- the degeneration of multiple tissues. Because the reprogramming of cells ming75. This point also introduces an additional important consideration. to an induced pluripotent state is accompanied by the induction of the Before a given iPS-cell disease model can be claimed to be truly represent- gene encoding telomerase reverse transcriptase (TERT), we investigated ative of the disease, how many patients must be involved, and how many whether the telomerase defect would limit the derivation and mainte- iPS cell lines must be derived from each patient? Although the answers to nance of iPS cells from individuals with dyskeratosis congenita. Although these questions are unclear, it is crucial to keep these issues in mind when the efficiency of iPS-cell derivation was poor, we were able to successfully generating disease models and making claims based on these models. reprogram patient . Surprisingly, whereas the mean telomere Although iPS cells are an invaluable tool for modelling diseases in vitro, length immediately after reprogramming was shorter than that of the the goal of developing patient-specific stem cells has also been motivated parental population, continued passage of some iPS cell lines by the prospect of generating a ready supply of immune-compatible cells led to telomere elongation over time. This process was accompanied by and tissues for autologous transplantation. At present, the clinical trans- upregulation of the expression of TERC, which encodes the RNA subunit lation of iPS-cell-based cell therapies seems more futuristic than the in of telomerase. vitro use of iPS cells for research and drug development, but two ground- Further analysis established that TERT and TERC, as well as DKC1, breaking studies have provided the proof of principle in mouse models were expressed at higher levels in dyskeratosis-congenita-derived iPS cells that the dream might one day be realized. Hanna, Jaenisch and colleagues than in the parental fibroblasts73. We determined that the genes encoding used homologous recombination to repair the genetic defect in iPS cells these components of the telomerase pathway — including a cis element derived from a humanized mouse model of sickle-cell anaemia76. Directed in the 3ʹ region of the TERC locus that is essential for a transcriptionally differentiation of the repaired iPS cells into haematopoietic progenitors active chromatin structure — were direct binding targets of the pluri- followed by transplantation of these cells into the affected mice led to potency-associated transcription factors. Further analysis indicated that the rescue of the disease phenotype. The gene-corrected iPS-cell-derived transcriptional silencing owing to a 3ʹ deletion in the TERC locus leads to haematopoietic progenitors showed stable engraftment and correction of the autosomal dominant form of dyskeratosis congenita by diminishing the disease phenotype. TERC transcription. Although telomere length is restored in dyskeratosis- In another landmark study from Jaenisch’s research group, Wernig congenita-derived iPS cells, differentiation into somatic cells is accompa- and colleagues derived dopaminergic neurons from iPS cells that, when nied by a return to pathogenesis with low TERC expression and a decay in implanted into the brain, became functionally integrated and improved telomere length. This finding showed that TERC RNA levels are dynami- the condition of a rat model of Parkinson’s disease77. The successful cally regulated and that the pluripotent state of the cells is reversible, sug- implantation and functional recovery in this model is evidence of the gesting that drugs that elevate or stabilize TERC expression might rescue therapeutic value of pluripotent stem cells for cell-replacement therapy defective telomerase activity and provide a therapeutic benefit. Although in the brain — one of the most promising areas for the future of iPS- we set out to understand the pathogenesis of dyskeratosis congenita with cell applications.

19 JANUARY 2012 | VOL 481 | NATURE | 301 © 2012 Macmillan Publishers Limited. All rights reserved Human pluripotent stem cell services Cell derivation and characterization: • IPSC generation and characterization • hES/hiPS directed differentiation into specialized cell types • hES/hiPS maintenance, expansion and banking

Genome engineering: • Genome editing of mammalian cells (hESC, iPSC, CSC) • Engineering effector molecules for gene regulation

Training in stem cell culture techniques Source materials

Fibroblast Neural cells Hair follicle Urine cells Keratinocytes Keratinocytes T cells Fibroblast Blood Adipose stem cells Urine Mesenchymal stem cells Cord blood stem cells Availability Availability Neural stem cells Germline stem cells Easy to reprogram

Blood: easy to obtain and easy to reprogram Fibroblast: easy to obtain, frozen stock and expandable Urine: easiest to obtain, easy to reprogram, potential contamination Comparison of IPSC derivation technology

Lentivirus RNA Sendai virus

Efficiency Episomal vector

Adenovirus Protein Safety Method Integra5on Efficiency Cost Retrovirus Yes 0.001 – 0.01% ++ Len9virus Yes 0.001 – 0.01% ++ Adenoviral Possible 0.0001 – 0.001% ++++ Sendai virus No 0.01 – 1% ++++ Episomal Possible 0.0001 – 0.01% + mRNA No >1% ++++ Protein No 0.00001% ++++ PROTOCOL

Sendai virus Figure 1 | Overview of the TiPSC generation Blood sampling Reseeding Picking up infection Isolation of PBMCs onto feeder cells TiPS colonies protocol. PBMCs are activated for 5 d with IL-2 (24 h) and anti-CD3 antibody, and then transduced with Passage SeV expressing human OCT3/4, SOX2, KLF4 and with cell count c-MYC. TiPSC colonies emerge at 20–25 d after blood sampling.

IL-2 + anti-CD3 antibody + ESC condition (bFGF +) Day 0 Day 5 Day 6 Days 20–25 the successful reprogramming of mono- T cells before Activated Infected Culturing 15–17 TiPS cells nuclear blood cells . In these methods, activation T cells T cells on feeder cells mononuclear blood cells from donors or frozen samples were infected using retro- 15 16,17 Activation with virus or lentivirus to express four IL-2 and factors, human OCT3/4, SOX2, KLF4 and anti-CD3 antibody

c-MYC. In these studies, human T cell Oct3/4 Sox2 reprogramming into iPSCs was achieved, but the efficiency of reprogramming was Klf4 c-Myc extremely low (approximately 0.0008– Reprogramming factors 0.01%). Although these methods used less peripheral blood and did not require the pharmacological Experimental design pretreatment of patients, the problems of transgene genomic Blood sampling. Our protocol is focused on the simple procedure insertion and low reprogramming efficiency remained, pre- of peripheral venous blood sampling to obtain the donor cells, cluding their wide use in the clinical application of iPSCs. using a standard process. Patient somatic cells can then be easily Generating iPSCs with TS-mutated SeV easily erases residual and aseptically obtained from the blood sample. In our protocol, genomic viral RNA from the target cells10, and the method 1 ml of whole blood is sufficient to generate TiPSCs (Fig. 2a). is significantly more efficient (~0.1%) compared with those protocols in which iPSCs were generated from T cells with Derivation of activated T cells. Peripheral blood mononuclear retrovirus or lentivirus. cells (PBMCs) can be separated by a Ficoll gradient method from Human keratinocytes derived from plucked human hair have also heparinized whole blood samples (Fig. 2b). Although PBMCs been used as another less invasive method of obtaining iPSCs from contain lymphocytes and monocytes, activation with plate-bound patient cells4,18. However, in some cases, these reported methods anti-CD3 monoclonal antibody and IL-2 selectively proliferates require several hairs to obtain successful cell outgrowth of keratino- T cells, and clearly increases the proportion of T cells in the cytes. Dental has also been explored as a potential source of cultured PBMCs11. CD3 protein exists in the complex of TCR iPSCs19. However, although teeth are routinely removed in many proteins on the surface of T cells, and can therefore be used clinics and no further procedures are required with respect to the as a T cell–specific marker. Anti-CD3 antibody modulates the donor, it is generally difficult to routinely obtain patients’ dental TCR-CD3 complex to induce T cell proliferation and activation20, © All rights reserved. 2012 Inc. Nature America, tissues—with specific genetic or nongenetic diseases—for the pur- whereas IL-2 also activates general T cell signaling pathways pose of iPSC studies. In comparison with these outlined methods, and eventually promotes cytokine transcription, cell survival, our protocol involves harvesting only a small sample of peripheral cell-cycle entry and growth21. At day 5 of culture with anti-CD3 Timelineblood; in addition, T cell proliferation for does not neediPSC stochastic cell monoclonal derivation antibody and IL-2, CD3 + cells increased up to ~95% outgrowth. These are clear advantages for clinical application in of cultured PBMCs (Fig. 2c–e). With this culture method, users comparison with the methods reported in the past. can avoid using a fluorescence-activated cell sorter in which

Blood%draw% Sendai%virus% Pick%iPS% Isola4on%of%PBMCs%% %infec4on%(24%h)% Colonies% colonies% Before After Reseeding%onto% a b c d PBMCs day 0 PBMCs day 5 centrifuging centrifuging PBMCs day 0 PBMCs day 5 feeder%cells% emerge% 250 CD3-positive CD3-positive 200 200 cells 71.2% cells 95.9% Erythroblast+expansion++PBMCs%Expansion% 150 Initial 1 weeks 1-2 weeks Primary 2 days 150

Count 100 Day%9% Day%11%CryopreservationCount 100 Day%20D23% Day%0% 50 Day%29D35% iPSC%medium%+%50 Contact Cells500 µm 500 µm EM%medium% Cytokines% 0 iPSC%medium% 0 hES%medium% 102 103 104 105 102 103 104 105 the Core FITC-A FITC-A Figure Patient2 | IsolationCCM#pa&ents# and sample activation of PBMCs. (a) Whole blood is collected into a 2.5-ml syringe by venipuncture.Methods: (b) Before e 100 centrifugation, two distinct layers are distinguishable in the 15-ml tube.Reprogramming The upper, dark red layer is the diluted blood and the 80 IRB lower layer is the Ficoll solution. After centrifugation, three distinct layers are apparent. The upper yellow layer containsSendai platelet- virus60 rich plasma, whereas the bottom clear layer is the Ficoll solution, and the thin white layer in between contains PBMCs (arrow). 40 (c) Morphology of PBMCs shortly after being seeded in the culture plate3-4 and activatedweeks using anti-CD3 antibody with IL-2 for 5 d. cells (%) CD3-positive 20 Tissue source: Proliferated T cells show many clusters in the culture plate at day 5 of activation. (d,e) Flow cytometric analysis Self-replicatingof isolated PBMCs RNA vector + 0 and PBMCs activated for 5 d with anti-CD3 antibody and IL-2 gated on the CD45 cell population. CD3 surface expressions of these 035 Blood, skin, hair populations were examined. The graph represents an average of three independent examinations. Error bars showProtein means ± s.d. Day

iPSCs NATURE PROTOCOLS | VOL.7 NO.4 | 2012 | 719

8-12 weeks 5-15 weeks

2-7 weeks Karyotyping Methylation Stem cell markers MicroRNA profiling Direct Teratoma formation Genome engineering differentiation EB differentiation into specialized cell types © 2012 Nature America, Inc. All rights reserved. less peripheral blood and did not require the pharmacological pharmacological the require not did and blood used peripheral less methods these Although 0.01%). 0.0008– (approximately low extremely was reprogramming of efficiency the but achieved, was iPSCs into reprogramming these In c-MYC. SOX2, and OCT3/4, KLF4 human factors, virus frozen samples were infected using retro or donors from cells blood mononuclear nuclear cells blood the successful reprogrammingmono of blood sampling. c-MYC SeV expressing human and anti-CD3 antibody, and then transduced with protocol. PBMCs are activated for 5 d with IL-2 Figure 1 populations were examined. The graph represents an average of three independent examinations. Error bars show means ± s.d. and PBMCs activated for 5 d with anti-CD3 antibody and IL-2 gated on the CD45 Proliferated T cells show many clusters in the culture plate at day 5 of activation. ( ( rich plasma, whereas the bottom clear layer is the Ficoll solution, and the thin white layer in between contains PBMCs (arrow). lower layer is the Ficoll solution. After centrifugation, three distinct layers are apparent. The upper yellow layer contains platelet- centrifugation, two distinct layers are distinguishable in the 15-ml tube. The upper, dark red layer is the diluted blood and the Figure 2 past. the in reported methods the with comparison in application clinical for advantages clear are These outgrowth. blood; in addition, T cell proliferation does not need stochastic cell our protocol involves harvesting only a small sample of peripheral pose of iPSC studies. In comparison with these outlined methods, tissues—with specific genetic or nongenetic diseases—for the pur patients’dental obtain routinely to difficult generally donor, is it procedures to areno further respect the and clinics required with iPSCs cytes. Dental tissue has also been explored as a potential source of require several hairs to obtain successful cell outgrowth of keratino cells patient been used as another less invasive method of obtaining iPSCs from with cells T from generated lentivirus. or retrovirus were iPSCs those with which in compared (~0.1%) protocols efficient more significantly is cells target the from residual RNA erases viral genomic easily SeV TS-mutated with iPSCs. iPSCs of Generating application clinical the in use wide their cluding pre remained, efficiency genomic reprogramming low and transgene insertion of problems the patients, of pretreatment a c ) Morphology of PBMCs shortly after being seeded in the culture plate and activated using anti-CD3 antibody with IL-2 for 5 d. Human keratinocytes derived from plucked human hair have also . TiPSC colonies emerge at 20–25 d after 1 1 5 9 or lentivirus . However, although teeth are routinely removed in many in removed routinely are However,. teeth although

| | Overview of the TiPSC generation Isolation and activation of PBMCs. ( PBMCs. of activation and Isolation CCM#pa&ents# Reprogramming Reprogramming blood to Pa9ents 4,1 8 . However, in some cases, these reported methods methods reported these cases, some However,. in using a integration-free method

studies, human T cell cell T human studies, OCT3/4, SOX2 15–1 16,1 7 b . In these . In 7 centrifuging to express four four express to Before , KLF4

methods, Passage&0 and centrifuging iPSC colonies in Feeders a After ) Whole blood is collected into a 2.5-ml syringe by venipuncture. ( - - Isola4on%of%PBMCs%%

& 1 0 Blood%draw% ES+like&colonies , and the method method the and , Day%0% c Erythroblast+expansion++ PBMCs day PBMCs%Expansion% EM%medium% 0 500 - - - Isolation ofPBMCs

Blood sampling T cellsbefore µ &

+ activation m

cell population. CD3 surface expressions of these T cells, and clearly increases the proportion of T cells in the the in cells T of proportion the increases clearly and cells, T proliferates selectively IL-2 and antibody monoclonal anti-CD3 contain lymphocytes and monocytes, activation with plate-bound ( samples blood whole heparinized by can from be cells method (PBMCs) a separated gradient Ficoll Derivation of activated T cells. 1 ml whole blood is of sufficient to generate TiPSCs ( protocol, our In sample. blood the from obtained aseptically and easily be then can cells somatic Patient process. standard a using cells, donor the obtain to sampling blood venous peripheral of Blood sampling. Experimental design can avoid using a fluorescence a using avoid can of cultured PBMCs ( and IL-2,antibody monoclonal CD3 growth and entry cell-cycle pathways signaling survival, cell transcription, cell cytokine promotes T eventually and general activates also IL-2 whereas activation and proliferation cell T induce to the complex TCR-CD3 modulates used antibody be Anti-CD3 marker. therefore can cell–specific and T a as cells, T of surface the on proteins PBMCs cultured Day d Passage&3 , IL-2 +anti-CD3antibody e RNA vector %infec4on%(24%h)% 0 ) Flow cytometric analysis of isolated PBMCs anti-CD3 antibody Sendai%virus% Activation with PBMCs day with cellcount IL-2 and Day%9% Passage & Reprogramming factors iPSC Day 5 5 Sendai virus 500 Activated Oct3/4 Cytokines% Our protocol is focused on the simple procedure infection + Klf4 1 T cells (24 h) %medium%+% 1 Day%11% . CD3 protein exists in the complex of TCR TCR of complex the in exists protein CD3 . µ m Day 6 c-Myc Fig. 2c Sox2 Reseeding%onto% d

Count feeder%cells% NATUREPROTOCOLS 100 150 200 250 50 0 b 2 Infecte – T cells 1 ) Before 10 e . At day 5 of culture with anti-CD3 anti-CD3 with culture . of 5 At day iPSC ). With this culture method, users users method, culture ). this With PBMCs day 2 d - Peripheral blood mononuclear mononuclear blood Peripheral 10 FITC-A activated cell sorter in which which in sorter cell activated CD3-positive %medium% cells 71.2 3 ESC condition(bFGF+) iPSCs feeder-free

+ 10 Fig. 2 Fig. onto feedercells

cells increased up cells increased to ~95% 4 on feedercell Reseeding 0 10 Culturing %

5 | b VOL.7 NO.4VOL.7 iPS ). Although PBMCs PBMCs Although ). Passage 12 Count 100 150 200 50 e 0 Colonies% Day%20D23%

CD3-positive s PROTOCOL emerge% cells (%) 10 100 20 40 60 80 PBMCs day 2 0 Fig. 2 TiPS colonies | 10 Days 20–25 FITC-A TiPS cell 2012 Picking up CD3-positive 035 3 cells 95.9 cells 10 Day a hES ). 4 | s 5

10 719 %medium% % 2 5 0 ,

colonies% Pick% Day%29D35%

iPS % Quality assurance: characterization

Immunofluorescence of of iPSCs pluripotency markers 14JL2 +/&+ 46,XX iPSC1&CCM3Karyotype

Alkaline phosphatase OCT4

NANOG SOX2

Teratoma forma9on SSEA-4 TRA-1-61 qRT-PCR of pluripotency markers hESCs% iPSCs% PBMCs% 1.6" 1.4" 1.2" NANOG% 1" SOX2%% 0.8" POU% 0.6"

Fold"change"" 0.4" 09July2014

relaIve"to"H7"WT" 0.2" 0" +/%' +/%' +/%' +/%' +/%'

38ED" H7"WT" 24SM" 40EG1"26CA3" 14JL2" 30SR7" H7%hES'iPSC%C1'iPSC%C2' iPSC%C3'25DN5"iPSC%C4' 26CA:PBMCs"14JL:PBMCs" iPSC2%CCM3iPSC3%CCM3iPSC1%CCM3 PBMCs1%CCM3PBMCs3%CCM3 Adriana Beltran, Bryan Richardson Differentiation of hES and IPS into three germ layers NESTIN SMA FOXA2

PAX6 CDX2 SOX17

Review Trends in Molecular Medicine August 2012, Vol. 18, No. 8

Specific cell type differentiation Somac cells Disease Pa5ents modeling Reprograming Neuronal differenSpecific cell type aon Reprogramming differen9a9on Drug Molecular discovery mechanism of the disease iPSCs ASD paent iPSC

Advances in genecs Cell and genomics therapy

Mesoderm Gene engineering Monogenic disorders: Ectoderm Endotherm Re syndrome, Idenficaon of novel Fragile X syndrome gene causing ASD Mesenchymal Timothy syndrome, etc. stem cells Hematopoie9c Pancrea9c Epithelial stem cells Animal progenitors stem cells Cardiac model Neural Progenitors Intes9nal Hepatocyte Progenitors stem cells progenitors

TRENDS in Molecular Medicine

Figure 1. Overview of adopting induced pluripotent stem cell (iPSC)Skin technology in autism spectrum disorderHemangioblast(ASD) research. ASD is a genetic neurodevelopmental

disorder. Monogenic syndromic ASDs, including Rett syndrome, Fragile X syndrome, or Timothy syndrome, are caused by well-defined genetic defects. AdvancedBeta cells

Pigment cells Immature

genomics tools are used to actively seek novel ASD genes. From skin biopsy or blood samples,Cardiomyocytes patient-derived ASD iPSCsEndothelial can be generated. Cortical neurons, which are

Astrocytes lung cells

most relevant neurons for ASD, canForebrain be directed from iPSCs using well-established protocols (Table 1). The neuronal cellscells can be used to elucidate novel disease

Hepatocytes

pathogenesis and to screen drugs asMidbrain potential treatments.Oligodendrocytes Recently developed gene editing technologies facilitate correcting the given mutation to provide isogenic

controls for in vitro disease modeling orSplinalcell-based therapy.

pluripotent they can give rise to all three germ layers (Box with ASDs, such as neuronal connectivity, synaptic activ-

1). Owing to their pluripotent and self-renewing capacities, ity, and neuronal migration, can be used as a screening

ESCs are considered cellular resources for modeling platform to test the efficacy of chemicals for ameliorating

human diseases as well as treating damaged human ASD.

organs and tissues [14–17]. iPSCs have been created by When using iPSCs as a cellular model for ASD, the

reprogramming differentiated cells by the ectopic expres- proper selection of standard control determines the inter-

sion of the four transcription factors Oct4, Sox2, c-Myc, and pretation of the in vitro phenotypes. Most often, iPSCs from

Klf-4 [18,19]. iPSCs and ESCs share characteristics such as healthy persons have been used as controls [21]. Because

morphology, expression of surface markers, pluripotent individual genetic variations have a large influence on

differentiation potential, and the capacity for self-renewal. cellular physiology, the use of iPSCs derived from closely

Because iPSCs retain the genetic composition of the par- genetically related persons, such as siblings or parents, can

ental somatic cells, the phenotypes of cells differentiated reduce the compounding genetic effect. Nevertheless,

from iPSCs presumably represent those manifested by isogenic iPSC lines are the ideal control. One method for

parental cells [20–25]. Thus, iPSCs from patients enable generating isogenic lines is to take advantage of the fact

the development of disease-specific cellular models, plat- that when a disease is X-linked and prominent in females,

forms for drug screening, and autologous sources for cell reprogramming produces unique sets of isogenic female

replacement therapies. Furthermore, iPSCs bypass the iPSCs by retaining the active/repressed X chromosome

ethical issues associated with destroying embryos to obtain status of fibroblasts [27]. Thus, iPSCs can be produced

human ESCs and immunological barriers that prevent the with either the wild type or mutant allele on the active

use of heterologous cells [26]. Figure 1 depicts a scheme for X chromosome from female Rett syndrome patients by

studying ASD using genomics, animal models, and iPSCs. taking advantage of this feature (see below and Box 2)

When a person is diagnosed with ASD, genomics methods [28–31]. However, a recent study showed that the inactive

can identify the culprit candidate genes and the physiolo- X chromosome in female iPSCs undergoes erosion of

gical functions of the genes can be examined by transgenic inactivation over time in culture, raising a concern for

murine models [9]. Meanwhile, iPSCs are readily available modeling X-linked disorders using iPSCs [32]. As gene

by reprogramming somatic cells from ASD patients. Fol- editing technologies such as zinc finger nuclease (ZFN)

lowing neuronal differentiation, functional analysis can be or transcription activator-like effector nucleases (TALEN)

performed to examine in vitro phenotypes manifested by have improved, manipulating single genes within iPSCs

ASD iPSCs. Quantifiable measures that are associated has become possible (Box 3) [22,33]. Gene editing technology

464 Neural differentiation timeline

Forebrain 15 days 35 days Neurons Midbrain Dopaminergic Spinal neurons 14 days ~90% Neural 10 days 14 days iPSC Progenitor 14 days Astrocyte Astrocytes Cells Precursor

14 days

Glial 40 days Myelina9ng Precursor Oligodendrocytes Oligodendrocytes +$Y27632$ 1$Y27632$ Day%% Derivation Derivation of functional endothelial

CD31 01%

CD31%%(PECAM+1)%(PECAM-1) VEGF0A% BMP4% FGF02% PLKO$ Ac(vin 0% %%A% Mesodermal%and%vascular%specifica(on% 1% P0## P1 P1 Suspension% CD144 (VE-cadherin) CD144 2% CD144%%(VE+cadherin)% 10X$ cells cells from 4% P2# P3 P3 SB431542% CCM3$ Adherent% 7% EC%amplifica(on% 14% ~70% Isola(on% iPSCs Tube forma5on assay PLKO$

CD31 Flow cytometry analysis Flow ofanalysis cytometry

CD144 (VE-cadherin) CD144 (PECAM-1)

CD31/CD144 ECs ECs CD31/CD144 20X$ CCM3$ Maintenance, expansion and banking of hES and hiPS cells

Embryonic stem cell lines distributed by the core: H1, H7 and H9 iPSC control lines

IMPORTANT: hES lines acquired from the core must be checked for pluripotency every month and be karyotyped every 20 passages. Human pluripotent stem cell services Cell derivation and characterization: • IPSC Reprogramming and characterization • hES/hiPS Differentiation • hES/hiPS Maintenance, expansion and banking

Genome engineering: • Genome editing of mammalian cells (hESC, iPSC, CS) • Engineering effector molecules for gene regulation

Training in stem cell culture techniques

Fast and easy genome engineering with CRIPSR/Cas9

23 bp genomic target sequence on January 4, 2013

DSB NHEJ HR repair

GFP

Gene of Gene of www.sciencemag.org interest interest

GFP

Knockout Knock-in Fig. 1. Genome editing in human cells using an engineered type II CRISPR system. (A) RNA-guided gene targeting in human Downloaded from cells involves co-expression of the Cas9 protein bearing a C terminus SV40 nuclear localization signal with one or more guide RNAs (gRNAs) expressed from the human U6 polymerase III promoter. Cas9 unwinds the DNA duplex and cleaves both strands upon recognition of a target sequence by the gRNA, but only if the correct protospacer-adjacent motif (PAM) is 20GG can in principle be targeted. (B) A genomically integrated GFP coding sequence is disrupted by the insertion of a stop codon and a 68bp genomic fragment from the AAVS1 locus. Restoration of the GFP sequence by homologous recombination (HR) with an appropriate donor sequence results in GFP+ cells that can be quantitated by FACS. T1 and T2 gRNAs target sequences within the AAVS1 fragment. Binding sites for the two halves of the TAL effector nuclease heterodimer (TALEN) are underlined. (C) Bar graph depicting HR efficiencies induced by T1, T2, and TALEN-mediated nuclease activity at the target locus, as measured by FACS. Representative FACS plots and

/ http://www.sciencemag.org/content/early/recent / 03 January 2013; / Page 4/ 10.1126/science.1232033

Genome editing possibilities

Genome editing using CRISPR/Cas9 based technologies are applied to any mammalian cell type (hES, hIPS and cancer cell lines).

• CRISPR-mediated gene knock-out.

• CRISPR-mediated gene knock-in.

• CRISPR-mediated point mutation introduction and/or repair to create isogenic cell lines. Genome editing timeline

Quality Contact with the Prepara5on Genera5on assurance core 2 weeks 8 to 12 weeks 2 weeks

Mycoplasma tes9ng Sequencing Design Transfec9on of cells of donor line Karyotype Experiments Selec9on and Mycoplasma tes9ng screening CRISPR design Create a

9meline Valida9on of posi9ve Reagent prepara9on clones CRIPSR mediated gene KO Editing the CCM3 locus

CCM3

Cerebral Cavernous Malforma0on (CCM) RhoGEF

RhoAŸGTP RhoAŸGTP

RhoGAP

RhoAŸGTP ROCK Y-27632

Ac5n stress fiber forma5on and loss of endothelial func5on

Control CCM3 loss CRIPSR mediated gene KO

5’ UTR Coding region 3’ UTR

1 2 4 5 6 7 8 9 10

Ex4A Ex8A

H7 hESC

iPSC T7 PCR 10X$ 20X$ PLKO$ CCM3$ PLKO$ CCM3$

1$Y27632$

+$Y27632$ +Y27632 CRIPSR mediated gene KO High efficient double allele KOs

Gene1

5’ UTR Coding region 3’ UTR

7 Gene1 1 2 3 4 5 6 9

Loading 2GuA control 1GuA 20% efficiency

Dele5on

Gene2 3’ Coding region 5’ UTR UTR 4 3 5 2 1 Gene2

Loading Ex5GuA Ex2GuA control Dele5on 40% efficiency CRIPSR mediated gene KO deletion of large genomic regions

Zawistowski et al. Cancer Discovery 2017 CRIPSR mediated gene KI Fluorescent reporter

CCM-YPET reporter real-5me imaging CCM-YPET fusion protein cellular distribu5on mimic CCM protein HCMEC-CCM-Yept

Felix Olivares and Adriana Beltran Engineering effector molecules for gene regulation

Ar0ficial Transcrip0on Factors (ATFs) Synthe0c transcrip0on factors Activator VP64 Domain A tumor suppressor (Maspin)

Repressor SKD Domain An oncogene (Sox2)

DNMT3A Chromatin Modifying An oncogene Domain (Sox2) non-methylated methylated MDA-MB-468 MDA-MB-231

Maspin Tubulin

Soft-agar assay Invasion assay Control ATF - DOX + DOX CONTROL CONTROL IF of ATF regulated gene (red) Tumorsphere assay - DOX + DOX DOX removal Control ATF ATF-126 ATF-126

Beltran, A, et al, Oncogene 2006 Nucleic Acids Research, 2012, Vol. 40, No. 14 6727 from ATCC (American Type Culture Collection, a terminal hemagglutinin (HA) decapeptide tag. The Manassas, VA, USA) maintained at 37ÀC and 5% CO2. correct ZF-sequence of the obtained product was con- firmed by plasmid sequencing. ATF construction Retrovirus infection of MDA-MB-435s The ZF target sites within the SOX2 promoter were se- lected using the website www.zincfingertools.org (42). The The pMX retroviral vectors containing the SOX2-ATFs selection of three 18-bp target sites was based on the close were first co-transfected with the plasmid (pMDG.1) ex- proximity to the transcriptional start site and the high pressing the vesicular stomatitis virus envelope protein into 293TGagPol cells to produce retroviral particles. content of GNN-triplets in the target sequence. One TM ATF was designed to target an 18-bp sequence in the Transfection was performed using Lipofectamine SOX2 enhancer region 1, 4-kb upstream of the TSS (Invitrogen, Carlsbad, CA) as recommended by the manu- (Figure 1B). Specific primers were designed coding for facturer. For cell proliferation and soft agar assays, cells the amino acids in the recognition helix of the ZFs respon- were harvested 24 h after the last infection. For flow Published online 4 May 2012 Nucleic Acids Research, 2012, Vol. 40, No. 14 6725–6740 sible for the binding to the target sequence (Figure 1C). cytometry analysis, protein, and mRNA extraction, doi:10.1093/nar/gks360 The ZF proteins were generated by overlapping PCR transduced cells were harvested 48 h post-infection. as described (43), SfiI-digested fragments were subcloned siRNA transfection into the retroviral vector pMX-IRES-GFP-SKD, generating pMX-ZF552SKD, pMX-ZF598SKD, pMX- MDA-MB-435s breast cancer cells were transfected with ZF619SKD and pMX-ZF4203SKD.Targeted Each ATF silencingcontains either a ofSOX2 the-specific oncogenic siRNA pool (siGENOME transcription an internal SV40 nuclear localizationfactor signalSOX2 (NLS) andin breastD-011778-01-04), cancer an irrelevant (non-specific) siRNA Sabine Stolzenburg1,2, Marianne G. Rots1, Adriana S. Beltran2, Ashley G. Rivenbark2,3, A ZF6 ZF5 ZF4 ZF3 ZF2 ZF1 SKD Xinni Yuan2, Haili Qian4NLS, Brian D. Strahl3,5 and Pilar Blancafort2,5,* HA-tag Nucleic Acids Research, 2012, Vol. 40, No. 14 6735 5’ 3’ 1 3’ Epigenetic Editing, Department of Pathology and5’ Medical Biology, University Medical Center Groningen, 2 UniversityZF-4203 of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands, Department of Pharmacology, x ATTAAGAGAGGAGGGGAA o x x 3 A B D o o B Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA, + -D +D ZF-598SKD –Dox or 4 SOX2 Core Promoter Region t D D 3 c K e S K State Key Laboratory of Molecular Oncology, Cancer Hospital/Institute, Chinese Academy ofZF-598SKD Medical +Dox v 8 8S TSS ty 9 9 5 p -5 -5 Sciences, Pan Jia Yuan Nan Li 17, Chaoyang District, Beijing 100021, P.R. China and UNC Lineberger F F -619 -598 -552 em Z Z Comprehensive-4203 Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA Regulatory Region 1 Regulatory Region 2 Received January 17, 2012; Revised March 22, 2012; Accepted April 11, 2012 Dox Induction tumor mm tumor volume AGCCTCCTCCCCCTCCTC GCCCCCTCCTCCCCCGGC GGCCCCGCCCCCTTTCAT ABSTRACTZF-619 ZF-598 ZF-552 INTRODUCTION OPEN Oncogene (2015) 34, 5427–5435 The transcription factor (TF) SOX2 is essential for Transcription© 2015 Macmillanfactors Publishers(TFs) Limited are All rights crucial reserved molecules 0950-9232/15 orches- C the maintenanceZF6 ZF5 of pluripotencyZF4 andZF3 self-renewalZF2 tratingwww.nature.com/oncZF1 gene programs involved in self-renewal, differenti- ation and organism’sC developmentalp=0.269 patterning. p=0.015 D ZF-552SKDin embryonicRRDELNV Q stemRANLR cells.A RSD InKLV additionR DPG toHLV itsR normalR SDK LVR D CRDLAR 150 Maintaining the proper threshold3 of expression of TFs is 2.5 ZF-598SKDstemDCR cellDLAR function,RSDK LVSOX2R QRAover-expressionHLER QRAHLER isR asso-SDK LVR D PGHLVR critical for the normal homeostatic function of cells and 2.0 ZF-619SKDciatedORIGINALRSDN withLVR cancer ARTICLERSDN LV development.R RSD KLVR TheRSDN abilityLVR toR SD se-N LVR T SGELVR tissues. Aberrant regulation of100 TF expression is frequently ZF-4203SKDlectivelyHKNALQ targetN R thisKDN LK andN otherQLAHLR oncogenicA QRAHLE TFsR inR cells,SDK LVR foundQSSN LV inR human malignancies and associated with specific 1.5 however,Stable remains oncogenic a significant challenge silencing due toin the vivotumorby subtypes programmable (1). Over-expression of oncogenic and TFs is 1.0 ‘undruggable’ characteristics of these molecules. well documented in the mammary50 gland, particularly in s D 5 1 8 3 SOX2 relative Here, we employ3 3 a zinc6 finger (ZF)-based artificial5 poorly differentiated, triple negative breast cancers 0.5 *

targeted de novo DNA methylation in breasttumor volume mm cancer -4 -2 4 4 expression mRNA A - - 2 B B B 2 1 -3 9 B 9 (TNBCs)Programmable (2). TNBCs oncogenic are characterized silencing in breast by cancer the lack of ex- TF (ATF)1 M approachM 9 toM selectively0 - suppressR 4 M SOX25 - - 1,2- 4 - 2 1 7 5 3 1 - 1 4 1 1,2 0 0.0 S StolzenburgF A A , AS5 BeltranA M, T Swift-ScanlanF -7 -B ,M AG RivenbarkA M ,Programmable R Rashwanpressionand ofoncogenic P Blancafort Estrogen silencing Receptor in breastS Stolzenburg (ER cancerÀ),et Progesterone al geneC expressionD D T inD cancerU C cells.R K WeU engineeredD U M M M B M S M Z S S M S Dox - + - + Dox - + - + S StolzenburgReceptoret (PR al À) and Epidermal Growthr Factorr Receptor r r four different proteins each composed5430 of 6ZF o o D D o o D D t t K K t t K K a-SOX2 2 (Her2À). Recent progress revealedec thatec some TNBCs ec ec v v 8S 8S v v 8S 8S arraysWith designed the recent comprehensive to bind 18 bp mapping sites of in cancer the genomes,SOX2 there is now a need for functional approachesy y to edit the9 aberrant9 5429 y y 9 9 belonging to the basal-liket and claudin-lowt -5 intrinsic-5 t t -5 -5 Binding p p F F Amplicon II p p F F a-Tubulinpromoterepigenetic and state enhancer of key cancer region, drivers to which reprogram controls the epi-pathology of the disease. In this studyem we utilizedem a programmableZ Z em em Z Z -654II -279 subtypes of breast cancerssite are highly aggressive and resist- SOX2DNA-bindingmethylation. methyltransferase The 6ZF to domains induce targeted were incorporation linked of DNA methylation (DNAme) in the SOX2 oncogene in breast -279 -1069 I -623 fi ATG +695III +1055 ant to treatment1.5 (3–5). It has-654 been proposed that these Figure 1. Design of ATFs to down-regulateto thecancerSOX2 Kruppel throughexpression. a Associated six zinc (A) Schematicnger (ZF)Box representation protein (SKD) linked repressor ofto a 6 DNA ZF ATF methyltransferase boundbreast to cancers DNA 3A with (ZF-DNMT3A). are the enriched orientation We in of demonstratedstem cells, which long-lasting might be the domains depicted. (B) Schematic5’ illustrationoncogenic of repression, the SOX2 whichpromoter was outlining maintained the even ZF-552SKD, after suppression3’ ZF-598SKD, of ZF-DNMT3A ZF-619SKD expression and ZF-4203SKDE in tumor cells.ZF-598SKD The de novo -DoxDNAme ZF-598SKD +Dox domain. Three engineered proteins were able to critical for tumor initiation,MCF7 progression and resistance to targeted sequences and their location3’ was relative faithfully to the propagated transcription and start maintained site (TSS). Highlightedthrough cell are generations5’ the core promoter even after (red), the regulatory suppression region of the 1 expression of the chimeric (green), and regulatory region 2 (blue).bind Arrows their show endogenous the orientation target of the 18-bp sites binding and effectivelysite in the promoterchemotherapy (from 50 to 31.00). (C and) Alpha-helical radiation ZF (6–11). Albeit their funda- methyltransferase in the tumor cells. Xenograft studies in NUDE mice demonstratedmRNA stable SOX2 repression and long-term breast amino acid sequences chosen to constructsuppress the ATFs.SOX2GCCCCCTCCTCCCCCGGC Residuesexpression at position (up –1, to+3 95%and +6 repression making specific contactsmental with role the recognition in tumor triplets etiology are and progression, TFs are tumor growth inhibition, which lasted for 4100 days post implantation of the tumor cells in mice. This was accompanied with a indicated in color (red refers to positionefficiencies) –1, blue to position in breast +3 and green cancer to +6 of cells. the ZF recognition Targeted helix). (Dcurrently) Quantification refractory of SOX2 expression to target-based drug discovery in 12 breast cancer cell lines by westernfaithful blot. maintenance of DNAme in the breast cancer implants. In contrast, downregulation of SOX2ZF598-by ZF domains engineered with linker approaches SOX2 0.5 due to their lack of small molecule binding 40x expression 40x down-regulationthe Krueppel-associated of SOX2 box repressorexpression domain resulted in a in transient and reversible suppressionDNMT3A of oncogenic gene expression.* Our decreased tumor cell proliferation and colony for- pockets.rel. Thus, novelfi strategies*** are required to** efficiently results indicated that targeted de novo DNAme of the SOX2 oncogenicsilence promoter the aberrant was suf expressioncient to induce of oncogenic long-lasting TFs epigenetic in cancer mation in these cells. Furthermore, induced expres- fi silencing, which was not only maintained during cell division butcells. also signi Ideallycantly0.0 these delayed novel the approaches tumorigenic phenotype should restore of cancer and cells in vivo, even inZF the absence of treatment. Here, weDNMT3A outline a genome-basedDOX targeting - + approach - + to long-lasting R - +tumor R growth sion of an ATF in a mouse model inhibited breast stably maintain the expressionZF598- pattern of these TFs, like inhibition with potential applicability to many other oncogenic drivers that are currentlyDNMT3A refractory E74A to drug design. cancer cell growth. Collectively, these findings it is observed in normal epithelial cells. demonstrate the effectiveness and therapeutic The SOX2 gene encodes a TF belonging to the 10x 10x Oncogene (2015) 34, 5427–5435; doi:10.1038/onc.2014.470; published online 16 February 2015 potential of engineered ATFs to mediate potent high-mobility groupZF598-SKD (HMG)ZF598-SKD ZF598-SKD family (12). SOX2 expression Figureempty 6. emptyvectorA SOX2 vector-specific ATF inhibits the growthAmplicon of pre-existing II s.c xenografts of MCF7 cells. (A) Time course plot of tumor volume monitored and long-lasting down-regulation of oncogenic TF is criticalby caliper for the measurements. maintenance AnimalsZF598-DNMT3A of self-renewalZF598-DNMT3A (NZF598-DNMT3A= 6) were in embryonic either maintained in a Dox-free diet (-Dox) or induced with Dox diet (arrow) at day 21 expression in cancer cells. stem cellspost-injection. (ESCs)-654 (B and) Picture neural of representative progenitor tumors cells collected (13–15). at day 28 post-induction from induced-279 empty vector, un-induced ZF-598SKD, and INTRODUCTION whereinduced it maintains ZF-598SKD self-renewal. animals.8,9 (CDNAme) Tumorin volume the SOX2 measurementspromoter at day 21 post-induction from empty vector and ZF-598SKD groups (N = 6 animals per group). Differences between groups were assessed by a Wilconxon rank sum test. (D) Quantification of SOX2 mRNA expression by qRT-PCR in Cytosine methylation in mammalian DNA is regarded as a key and enhancer regions functions as an epigenetic switch, which tumor samples from a representative tumor xenograft. Bar10 graphs represent the mean and SD of three tumorno Dox samples. Differences in gene expression epigenetic modification controlling essential processes such as forceswere cells calculated to activate with multiple a Student’s differentiationt-test, *P = pathways. 0.01 (E) Hematoxylin-EosinSOX2 is staining of representative ZF-598SKD –Dox and +Dox tumor sections. 1 15 empty vector 10–12 imprinting, silencing of retrotransposons and cell differentiation. thereforeUn-induced not expressed (–Dox) animals in most revealed normal highly adult compact tissues. tumors. Induced (+Dox) ZF-598SKD sections comprised discrete islands of tumor cells, *ToIn addition whomempty correspondence to vector DNAempty methylation vectorZF598-SKD shouldZF598-SKD be (DNAme), addressed.ZF598-SKD repressiveZF598-DNMT3A Tel: +1ZF598-DNMT3A 919 histone 966ZF598-DNMT3A 1615; post- Fax: +1Moreover, 919separated 966 aberrant5640; by Email: intervening reactivation [email protected] stroma. of PicturesSOX2 has were been taken detected at 10 and in a detail of a 40 magnification is shown. ZF598-DNMT3A À À translational modifications reinforce gene inactivation, resulting in ~ 43% of basal-like breast cancers and in several other malig- +Dox DOXÀ The Author(s) - +2012. Published - + by Oxford R University - Press. + R ZF598-SKD 2 nancies, including10 glioblastoma, lung, skin, prostate and ovarian Thisthe is formationan Open Access of articleinactive distributed chromatin, under namelythe terms heterochromatin.of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/ SOX2 37kDA 13 (R) by-nc/3.0),Importantly, which the permits epigenetic unrestricted information non-commercial must use, be distribution, stably trans- and reproductioncarcinomas. in any medium,SOX2 providedoverexpression the original in tumorwork is specimensproperly cited. has been mitted during mitosis for the proper maintenance of cellular associatedin tumors with derivedboth promoter from empty hypomethylation vector control relative (data to not accompanied by a decreased nuclear SOX2 staining HA-tag 3 42kDA fi 14,15 identity, a process referred as epigenetic memory. In pathological adjacentshown). normal5 In tissue contrast, and copy the number ZF-598SKD ampli cations. +Dox tumorsThe ex- (Figure 7A), and by a decreased proliferation of the states such as cancer, the epigenetic landscape of normal cells overexpression of SOX2 in breast cancer hasAmplicon been shown I to HA-tag 55kDA hibited-1069 a more organized tissue with increased amount tumor cells, as-623 indicated by a Ki67 staining of the becomes disrupted. Aberrant incorporation or removal of DNAme directlyofFold increase in activate interveningCYCLIN stroma D1, resulting separating in an increased small islands mitotic index of tumor tumor sections (Figure 7B). In summary, our in vivo in cancer cells leads to genome-wide alteration of gene and proliferation.16,17 The downregulation of SOX2 by RNA cells to t=0 release rel. ATP (Figure 6E, right panel). In addition, immunofluor- analyses indicated that the tumor suppressive functions H3expression, including inactivation of tumor suppressor genes17kDA interference decreased0 the tumorigenic phenotype in the lung, escence analyses of13,16,18 the tumor sections demonstrated that of the engineered silencers were maintained after and reactivation of oncogenes.4 Notably, changes in DNAme breast and ovarian0 cancers.48 However,96 a major limitation144 of no Dox patterns are characteristic hallmarks of the distinct intrinsic smallthe interferin ZF proteins RNA and small weretime hairpin expressed (hrs) RNA approaches in the nucleus in cancer of the long-term inoculation of the tumor cells, resulting in subtypes of breast cancer. Poorly differentiated, highly prolifera- therapymajority has been of tumor the short cells half-life in ZF-598SKD of small RNA +Dox or animals, the the maintenance of the SOX2 down-regulation and Figure 1. Stable downregulation of the SOX2 expression by ZF598-DNMT3A. (a) Schematicbut not in illustration un-induced of the animalsSOX2 (Figurepromoter 7A) indicating or controls the decreased tumor cell+Dox proliferation in animal models of tive basal-like breast cancers associated with stem/- methylation of the virally encoded small hairpinfi RNA promoter, domain structurelike of features the ZF598-DNMT3A are significantly hypomethylated construct, itsbinding relative to site the otherand thewhich three(data results amplicons not in transient shown). analyzed effects This byin induction sodium vivo. bisul of ZFte expression sequencing was or breast cancer. MassARRAYs (amplicon I, blue; II, purple and III, green). (b) Quantification of SOX2 mRNA expression by qRT-PCR in MCF7 cells. Cells were breast cancer subtypes. Importantly, these tumors are driven by In light of the essential role of DNAme in the regulation of SOX2 (R) stably transduced with empty vector, ZF598-SKD and ZF598-DNMT3A. The expression of the ZF fusion was controlled by addition or aberrant activation of multiple developmental transcription expression we hypothesized that targeted de novo methylationfi in removal of doxycyclinefactors (TFs), (Dox); which R = fuelDox the removal. tumor with Cells sustained were induced proliferation, with Doxthe everySOX2 48promoter h and either would harvested result in an at epigenetic 72 h after 'off'rst switch, induction (+Dox) or removeddrug from resistance Dox and and metastatic subcultured capacity. for5– 87 days and processed forforcing western cancer blot. cells Error to bars undertake represent differentiation standard deviation programs. (s.d.) (***P 0.001, **P 0.01, *P 0.05). (c) Detection of SOX2 by western blot. The C-terminal Hemagglutinin (HA)Amplicon tag was III used for o Theo High Mobilityo Group oncogenic TF SOX2 is normally Furthermore, because+695 DNAme is read and written by endogenous +1055 immunodetectionexpressed of the ZF in proteins. embryonic An stem anti-histone cells and neural H3 antibody progenitor was cells, used asproteins loading andcontrol. faithfully (d) Cell transmitted viability of during MCF7 cellscelldivision, assessed we by Cell TiterGlo assays. Empty vector, ZF598-SKD and ZF598-DNMT3A-transduced cells were induced with Dox for 48 h and removed from Dox after 72 h (red arrow). P-values between empty vector and ZF598-DNMT3A and between ZF598-SKD and ZF598-DNMT3A were Po0.001 and 1 no Dox Po0.05 respectively,Cancer at Epigenetics 144 h. Group, The Harry Perkins Institute of , Western Australia & School of Anatomy, Physiology and Human Biology, M309, The University of Western Australia, Nedlands, Western Australia, Australia; 2Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; 3Lineberger Comprehensive Cancer Center/School of Nursing, University of North Carolina, Chapel Hill, NC, USA and 4Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA. Correspondence: Professor P Blancafort, Cancer Epigenetics Group, The Harry Perkins Institute of Medical +Dox To evaluate theResearch, spreading Western Australia of DNAme & School in of the Anatomy,SOX2 Physiologylocus after and Human the Biology,could M309, The be University associated of Western with Australia, propagation 6 Verdun Street or Nedlands, spreadingNedlands, of Western DNAme Australia 6009, Australia. (R) initial induction,E-mail: we analyzed [email protected] two additional amplicons (amplicon further away from the 6ZF site during DNA replication. These I and III) flankingReceived the 6ZF-binding 14 July 2014; revised site, 15encompassing November 2014; accepted ~ 1 9 kbps December up- 2014; publishedresults online further 16 February support 2015 the increased downregulation of SOX2 and downstream of the translation start site (Figures 2c and d).Figure 2. expressionExpression of after the ZF598-DNMT3A Dox removal induces previously targeted observed DNA methylation by quantitative in the SOX2 promoter. (a) Sodium bisulfite-sequencing analysis Sequencing analysis of amplicon I (−1069 to − 623 bps upstreamof DNA derivedreverse from transcription MCF7 cells stably polymerase transduced chain with reaction empty vector, and ZF598-DNMT3A western blot and ZF598-DNMT3A-E74A mutant and induced with of the translation start site) demonstrated an increase of DNAmeDox. The analyzed amplicon II expands from − 654 to − 279 base pairs (bps) upstream the translation start site and includes the 6ZF-binding site (positionanalysis− 598 and relative outline to the the translation importance start site). of GrayCpG circles dinucleotides indicate notflanking analyzed methylation values owing to CpGs with high- or low- upon ZF598-DNMT3A expression as compared with the MCF7mass Daltonthe peakscore promoterfalling outside for the the conservative regulation window of SOX2 of reliableexpression. detection for the EpiTYPER software, colored circles indicate variously empty vector transduced cell line or ZF598-DNMT3A-E74A-methylated CpGs. (b) MassARRAY analysis of the same amplicon as in (a). Circles indicate the CpG dinucleotides in the amplicon (Color code: transduced cells. The frequencies of DNAme in amplicon I wereyellow = unmethylated CpG to blue = 100% methylated CpG). The percentage of meCpG was determined in MCF7 control-transduced cells fi and in MCF7 cells stably transduced with ZF598-DNMT3A or ZF-598-DNMT3A-E74A in absence of Dox (no Dox), presence of Dox (+Dox) or signi cantly higher than those immediately adjacent to the ZF-after DoxZF598-DNMT3A removal (R, eight days). expression (c) MassARRAY confers analysis anticancer of amplicon memory I (−1069 and to − 623 bps upstream of the translation start site) in Dox-free binding site, suggesting that DNAme was possibly spread andconditionsreduces (no Dox) tumor and after growth Dox induction in a breast (+Dox) cancer Dox removal xenograft (R). (d in) MassARRAY NUDE mice analysis of amplicon III (+695 to +1055 bps) downstream fl even reinforced in the anking regions. Most importantly, afterof the translationTo analyze start site, whether same conditions the ZF598-DNMT3A as above. construct induced discontinuation of the ZF598-DNMT3A expression (Dox removal), phenotypic memory in vivo we took advantage of our inducible the de novo methylation additionally increased up to 97% at MCF7 cell lines stably transduced with either the catalytically specific CpG sites in Amplicon I (Figure 1c). active ZF598-DNMT3A or the empty vector control. A total of As last, the MassARRAY analysis of DNAme in amplicon III (+695 6 Oncogene2 (2015)× 10 5427MCF7– 5435 cells transduced with either ZF598-DNMT3A or © 2015 Macmillan Publishers Limited to +1055 bps downstream of the translation start site, Figure 2d) fl revealed no methylation in MCF7 control and ZF598-DNMT3A cells control were implanted into the ank of NUDE mice and allowed in the absence of Dox. Upon ZF598-DNMT3A expression (+Dox) to grow for 22 days before switching the animals to a Dox- CpG methylation increased up to 80%, and no DNAme was containing diet (Figure 3a). Within each group N = 5 animals were induced in cells expressing the ZF598-DNMT3A-E74A mutant. Our maintained in a Dox-free diet. At day 19 post induction, half of the time-course analysis suggests that the de novo DNAme induced by ZF598-DNMT3A injected animals from the +Dox group were the artificial methyltransferase results in both, a phase of induction removed from the Dox diet (N = 10), to withdraw the expression of of gene silencing upon Dox induction, and a phase of the ZF methyltransferase (R). In contrast with ZF598-DNMT3A, maintenance and reinforcement of silencing (Dox removal), which catalytic dead ZF598-DNMT3A-E74A mutant cells failed to reduce

© 2015 Macmillan Publishers Limited Oncogene (2015) 5427 – 5435

Training in stem cell culture techniques

The Stem Cell Core Facility offers a one-to-one training course in basic human stem cell culture techniques.

• Introduc9on to stem cell biology • Morphology of hES and hiPSCs • Media prepara9on • Feeder prepara9on • Feeder-free ECM coa9ng • Passaging hES and hiPSCs by enzyme and manual picking • Freezing/thawing hiPSCs Available technology Two Leica stereomicroscopes housed in two Copper-lined CO2 QuantStudio™ 7 Flex EVOS FL cell biosafety cabinets incubators Real-Time PCR System EVOS xl Core imaging system

Countess® II FL Veri9® 96-Well Fast Automated Cell Qubit 3.0 Thermal Cycler Counter Neon Electroporator Fluorometer Going forward

Collabora9ve research Expand technological footprint Build long term rela9onships

The HPSC core is tightly coupled to the success of its users and collaborators Take home message

The Human stem cell core can support your stem cell studies by:

1. Reprogramming somatic cells into iPS cells. 2. Differentiate hES and iPS cells into specialized cell types. 3. Develop protocols for directed differentiation. 4. Design and engineer gene editing tools for: • Making isogenic iPS cells. • Gene knockouts. • Deletion of long enhancers and/or short regulatory elements. • Making fluorescent reporter lines. 5. Design and engineer synthetic transcription factors for gene regulation Thank you