Table of Contents Table of Contents
Previous Page  69 128 Next Page
Information
Show Menu
Previous Page 69 128 Next Page
Page Background

multiplex activation of the neurogenic factors

Brn2

,

Ascl1

, and

Myt1l

(BAM factors). We hypothesized that targeted activation

of the endogenous genes in PMEFs, as opposed to the forced

overexpression of the corresponding transgenes, could more

directly access the endogenous loci and rapidly remodel their

epigenetic signatures, thus potentially reflecting a more natural

mechanism of action and serving as an alternate method to

achieve cell lineage conversion.

In PMEFs, the

cis

-repressive chromatin landscape at neuronal

loci may preclude binding of regulatory factors, in turn impeding

transcriptional activation. As a result, expression of the BAM fac-

tors in PMEFs from exogenous vectors likely relies on stochastic

processes for reactivation of the corresponding endogenous

genes. Furthermore, transient delivery of the BAM factors, as

done in our initial experiments (Figures 1, 2, and 3), limits the

time window within which the endogenous networks and posi-

tive feedback loops can be established. We demonstrated that

targeting the endogenous genes directly induced the enrichment

of histone H3 modifications H3K27ac and H3K4me3 at the

Brn2

and

Ascl1

loci at 3 days post-transfection, whereas transgene

overexpression via transfection of plasmids encoding the re-

programming factors did not alter these chromatin marks (Fig-

ures 3 and S4). Additionally, we observed sustained high levels

of expression from the endogenous genes at later stages of re-

programming despite the transient delivery of the gRNA plas-

mids (Figure S3).

In contrast, we found that stable integration and constitutive

expression of the exogenous reprogramming factors via lentiviral

delivery led to the eventual deposition of H3K27ac at their endog-

enous loci with a concomitant improvement in reprogramming

capacity (Figures 4F and 4G). We did not observe a similar

improvement with constitutive expression of

VP64

dCas9

VP64

and

gRNAs, which is possibly attributable to the lower levels of overall

expression of the neuronal transcription factors achieved by

transactivation of the endogenous genes compared to ectopic

overexpression. Consequently, the direct activation of the endog-

enous genes via

VP64

dCas9

VP64

may be more amenable to tran-

sient delivery approaches that avoid undesired consequences

of vector integration into the genome. Such transient methods,

including the direct delivery of ribonucleoprotein Cas9-gRNA

complexes, may be a more clinically translatable method of

generating reprogrammed cells that are genetically unmodified.

Achieving robust and well-defined reprogrammed cell popula-

tions is still a central challenge. Reprogrammed cells often fail to

acquire completely mature phenotypes and can retain epige-

netic remnants of the native cell type (Kim et al., 2010). Moreover,

a recent study demonstrated that reprogramming efficiency can

be limited by divergence to a competing cell identity (Treutlein

et al., 2016). The molecular mechanisms and practical conse-

quences of these limitations are largely unknown. As the toolkit

of designer transcription factors expands to precisely modify

the epigenome (Hilton et al., 2015; Kearns et al., 2015; Maeder

et al., 2013a; Mendenhall et al., 2013; Thakore et al., 2016), these

tools may be used to prime specific genomic loci in diverse cell

types, promote endogenous transcription factor binding, and

directly correct regions of epigenetic remnants that prove to be

problematic for a given application. This may lead to improved

reprogramming fidelity and extension of the breadth of donor

cells amenable to reprogramming.

EXPERIMENTAL PROCEDURES

Cell Culture, Transfections, and Viral Transductions

PMEFs were maintained in high serum media during transduction and trans-

fection of expression plasmids and subsequently cultured in neurogenic

serum-free medium for the duration of the experiments to promote neuronal

survival and maturation. Lentivirus was produced in HEK293T cells using the

calcium phosphate precipitation method. All transfections were performed us-

ing Lipofectamine 3000 (Invitrogen) in accordance with the manufacturer’s

protocol. All expression plasmids used in this study can be found in Table S2.

Immunofluorescence Staining and qRT-PCR

All sequences for qRT-PCR primers can be found in Table S3. Total RNA

was isolated using the QIAGEN RNeasy and QIAshredder kits, reverse tran-

scribed using the SuperScript VILO Reverse Transcription Kit (Invitrogen),

and analyzed using Perfecta SYBR Green Fastmix (Quanta BioSciences). All

qRT-PCR data are presented as fold change in RNA normalized to

Gapdh

expression. For immunofluorescence staining, cells were fixed with 4% para-

formaldehyde, permeabilized with 0.2% Triton X-100, and incubated with pri-

mary and secondary antibodies.

Electrophysiology

A synapsin-RFP lentiviral reporter was used to identify cells in co-culture with

primary rat astroglia for patch-clamp analysis at indicated time points. Action

potentials and inward and outward currents were recorded in whole-cell config-

uration. Data were analyzed and prepared for publication using pCLAMP and

MATLAB.

Chromatin Immunoprecipitation qPCR

Chromatin was immunoprecipitated using antibodies against H3K27ac and

H3K4me3, and gDNA was purified for qPCR analysis. All sequences for

ChIP-qPCR primers can be found in Table S3. qPCR was performed using

SYBR green Fastmix (Quanta BioSciences), and the data are presented as

fold change gDNA relative to negative control and normalized to a region of

the

Gapdh

locus.

Mouse ENCODE ChIP-Sequencing Datasets

H3K4me3 and H3K27ac ChIP-sequencing data from C57BL/6 E14.5 whole

brain and mouse embryonic fibroblasts (GSE31039) were acquired from the

Mouse ENCODE Consortium generated in Bing Ren’s laboratory at the Ludwig

Institute for Cancer Research.

Statistical Methods

Statistical analysis was done using GraphPad Prism 7. All data were analyzed

with at least three biological replicates and presented as mean ± SEM. See

figure legends for details on specific statistical tests run and p values calcu-

lated for each experiment.

SUPPLEMENTAL INFORMATION

Supplemental Information includes Supplemental Experimental Procedures,

four figures, and three tables and can be found with this article online at

http://dx.doi.org/10.1016/j.stem.2016.07.001

.

AUTHOR CONTRIBUTIONS

J.B.B.,A.F.A.,K.W.L.,andC.A.G.designed experiments.J.B.B.,A.F.A.,H.-G.W.,

A.M.D., and H.A.H. performed the experiments. All authors analyzed the data.

J.B.B. and C.A.G. wrote the manuscript with contributions by all authors.

CONFLICTS OF INTEREST

C.A.G. and J.B.B. are inventors on filed patent applications related to this work.

ACKNOWLEDGMENTS

The authors thank Ami Kabadi, Pablo Perez-Pinera, and Syandan Chakraborty

for providing plasmid constructs; Christopher Grigsby for protocols for PMEF

Cell Stem Cell

19

, 406–414, September 1, 2016

413