Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Downloaded from https://academic.oup.

com/stmcls/article/30/4/673/6415666 by CINVESTAV user on 26 May 2022


Efficient Stage-Specific Differentiation of Human
Pluripotent Stem Cells Toward Retinal Photoreceptor
Cells
Carla B. Mellough, Evelyne Sernagor, Inmaculada Moreno-Gimeno, David H.W.
Steel, Majlinda Lako
EMBRYONIC STEM CELLS/INDUCED PLURIPOTENT STEM CELLS
Efficient Stage-Specific Differentiation of Human Pluripotent
Stem Cells Toward Retinal Photoreceptor Cells
CARLA B. MELLOUGH,a EVELYNE SERNAGOR,b INMACULADA MORENO-GIMENO,c
DAVID H.W. STEEL,a,d MAJLINDA LAKOa
a
Institute of Genetic Medicine; bInstitute of Neuroscience, Newcastle University, Newcastle, United Kingdom;
c
Centro de Investigacion Principe Felipe, Valencia, Spain; dSunderland Eye Infirmary, Sunderland,

Downloaded from https://academic.oup.com/stmcls/article/30/4/673/6415666 by CINVESTAV user on 26 May 2022


United Kingdom

Key Words. Embryonic stem cell • Photoreceptor • Retinal pigmented epithelium • Outer retinal degeneration

ABSTRACT
Recent successes in the stem cell field have identified some their ability to endogenously upregulate the expression of a
of the key chemical and biological cues which drive photo- range of factors important for retinal cell type specifica-
receptor derivation from human embryonic stem cells tion. However, cultures that were differentiated with full
(hESC) and human induced pluripotent stem cells (hiPSC); supplementation under our photoreceptor-induction regi-
however, the efficiency of this process is variable. We have men achieve this within a significantly shorter time frame
designed a three-step photoreceptor differentiation proto- and show a substantial increase in the expression of photo-
col combining previously published methods that direct receptor-specific markers in comparison to cultures differ-
the differentiation of hESC and hiPSC toward a retinal lin- entiated under minimal conditions. Interestingly, cultures
eage, which we further modified with additional supple- supplemented only with B27 and/or N2 displayed compara-
ments selected on the basis of reports from the eye field ble differentiation efficiency to those under full supplemen-
and retinal development. We report that hESC and hiPSC tation, indicating a key role for B27 and N2 during the
differentiating under our regimen over a 60 day period differentiation process. Furthermore, our data highlight an
sequentially acquire markers associated with neural, reti- important role for Dkk1 and Noggin in enhancing the
nal field, retinal pigmented epithelium and photoreceptor differentiation of hESC and hiPSC toward retinal progeni-
cells, including mature photoreceptor markers OPN1SW tor cells and photoreceptor precursors during the early
and RHODOPSIN with a higher efficiency than previously stages of differentiation, while suggesting that further
reported. In addition, we report the ability of hESC and maturation of these cells into photoreceptors may not
hiPSC cultures to generate neural and retinal phenotypes require additional factors and can ensue under minimal
under minimal culture conditions, which may be linked to culture conditions. STEM CELLS 2012;30:673–686
Disclosure of potential conflicts of interest is found at the end of this article.

complex underlying genetic etiology. In such cases, cell


INTRODUCTION replacement remains an important option.
A seminal paper by MacLaren et al. demonstrated that
Outer retinal degeneration (ORD) is the leading cause of the only cells capable of successful integration and differen-
blindness in the developed world. There is still no treatment tiation into functional rod photoreceptors in the mouse retina
for many forms of ORD where retinal pigmented epithelium were postnatal postmitotic cells that were already committed
(RPE) dysfunction and photoreceptor demise predominate. to a photoreceptor fate [6]. By contrast, only embryonic
Clinical trials indicate some protective effects of dietary sup- stage Crx-positive cells were able to differentiate into cone
plements or intraocularly injected trophic factors in slowing photoreceptors following transplantation [7]. For photorecep-
disease progression, but these are not a definitive cure. Gene tor replacement in humans, the equivalent donor cell type
therapy strategies for specific forms of hereditary retinal dys- would have to be obtained from the second trimester
trophy (HRD) are under development, and the viability of this embryo, which raises clear ethical concerns. A more viable
approach for partial restoration of visual function has been option would be to try and differentiate human embryonic
demonstrated [1–5]. Yet this approach remains ineffective in stem cells (hESC) and/or human induced pluripotent stem
cases where a substantial loss of photoreceptors has already cells (hiPSC) into photoreceptors in vitro prior to
occurred and is difficult to achieve in HRD resulting from transplantation.

Author contributions: C.B.M.: designed and performed research, analyzed data, wrote the paper, and approved final version of
manuscript; E.S. and D.H.W.S.: performed research and contributed to paper writing and final approval of manuscript; I.M.G.:
performed research, data writing, and final approval of manuscript; M.L.: designed and performed research and contributed to paper
writing, final approval of manuscript, and fund raising.
Correspondence: Majlinda Lako, M.Sc., Ph.D., International Centre for Life, Institute of Genetic Medicine, Newcastle University,
Newcastle NE1 3BZ, U.K. Telephone: 44-1912418688; Fax: 44-1912418666; e-mail: majlinda.lako@ncl.ac.uk Received July 7, 2011;
accepted for publication December 20, 2011; first published online in STEM CELLS EXPRESS January 20, 2012. V C AlphaMed Press

1066-5099/2012/$30.00/0 doi: 10.1002/stem.1037

STEM CELLS 2012;30:673–686 www.StemCells.com


674 Efficient Photoreceptor Generation from Human Stem Cells

Several key publications have shown that it is possible to [24]) were grown under atmospheric oxygen conditions on
drive the differentiation of hESC and hiPSC toward photorecep- mitotically inactivated mouse embryonic fibroblasts (MEFs) in
tors and RPE using various approaches ([8–15] and Supporting hESC medium containing knockout-Dulbecco’s modified
Information Table S1). It has been suggested that early retinal Eagle’s medium (DMEM), 1 mM L-glutamine, 100 mM nones-
progenitor cells (RPCs) can be obtained from ventral sential amino acids (NEAA), 20% knockout serum replacement
neural populations derived from differentiating hESC and hiPSC (KOSR, Gibco), 1% penicillin-streptomycin and 8 ng/ml bFGF
cultures in the absence of growth factors; however, this process (Invitrogen). hESC/hiPSC medium was changed daily and
is heavily dependent on the cell line-specific endogenous signal- hESC/hiPSC colonies were passaged every 4-5 days at a ratio of
ing, especially those related to Dkk1 and Noggin [11, 16]. 1:3-1:4 by incubation in 1 mg/ml collagenase IV. Prior to hESC/
The RPE, which can be generated during the spontaneous hiPSC differentiation, MEFs were removed by culture of hESCs
differentiation of hESC and hiPSC upon removal of basic fibro- and hiPSCs on hESC-qualified mouse matrigel (BD Biosciences,
blast growth factor (bFGF; Supporting Information Table S1), New Jersey) for two passages. Differentiation experiments were

Downloaded from https://academic.oup.com/stmcls/article/30/4/673/6415666 by CINVESTAV user on 26 May 2022


is critical to the normal function and survival of the light- initiated using the protocol outlined in Figure 1A. hESC/hiPSC
sensitive photoreceptors; indeed the two cell types are interde- clumps were harvested following collagenase treatment and cul-
pendent on each other. It is likely that in many forms of ORD, tured in low adherence bacteriological plates (Fisher Scientific,
the replacement of both cell types will be required, highlighting Loughborough, UK) to form embryoid body (EB) cultures. Con-
the need to identify new renewable cell sources that can be used trol cultures were differentiated in a basal ventral neural induc-
for restorative purposes. The restorative potential of hESC- and tion media (VNIM) (Fig. 1A, blue dashed line) consisting of
more recently hiPSC-derived RPE has already been demon- DMEM/Ham’s F-12 (DMEM/F-12, PAA, Linz, Austria) contain-
strated in the dystrophic Royal College of Surgeons (RCS) rat ing decreasing concentrations of KOSR (20% for the first 5 days,
[17–21]. RPE transplantation alone, however, although protec- 15% until day 9 then 10% until day 37) supplemented with L-glu-
tive for remaining photoreceptors, does not act to replace photo- tamine, NEAA, and B27 (1:50, PAA). On day 30 of differentia-
receptors that have already been lost. Additionally, the potential tion, EB suspension cultures were transferred to attachment cul-
of hESC-derived photoreceptors for cell replacement has been ture conditions on tissue culture were coated with poly-L-
demonstrated [22, 23], yet photoreceptor differentiation proto- ornithine (dissolved at 10 lg/ml in sterile dH2O, Sigma, Dorset,
cols remain variable and in some cases require extensive culture UK) and laminin (5 lg/ml in phosphate buffered saline [PBS],
periods (Supporting Information Table S1). Two studies [22, Sigma) for the remaining 30 days. On day 37, culture medium
23] have shown that hESC- and hiPSC-derived retinal cell was changed to a basal KOSR-free formula (Fig. 1A, blue
grafts can express photoreceptor markers and integrate within dashed line) consisting of DMEM/F12/L-glutamine/NEAA/B27
the outer nuclear layer in intact newborn and adult retina. How- and N2 (1:100, Gibco, Paisley, UK).
ever, following subretinal transplantation into Crx/ mice (an To drive photoreceptor induction from hESC/hiPSC (which
animal model of Leber’s Congenital Amaurosis), these cells we have termed PR-induced cultures, shown by the red dashed
failed to develop photoreceptor outer segments, indicating that line in Fig. 1A), hESC/hiPSC were differentiated as for control
their final maturation and functional integration within the com- cultures but with VNIM that was supplemented with recombi-
promised retina remains incompletely understood. Thus, devel- nant mouse (rm) Noggin (1 ng/ml, R&D Systems, Abingdon,
opment of protocols aimed at efficiently generating enriched UK), recombinant human (rh) Dickkopf-1 (Dkk1, 1 ng/ml, R&D
population of photoreceptor precursors at the correct ontoge- Systems), Insulin-like growth factor 1 (rhIGF-1, 5ng/ml, Sigma)
netic stage is key prior to any translation of cell-based restora- rhLefty A (500 ng/ml, R&D Systems), Human Sonic Hedgehog
tive therapies for the treatment of photoreceptor loss. (Shh, 30 nM, Peprotech, London, UK) and 3,30 ,5-triiodo-L-thyro-
With this aim, following on from previous reports nine (T3, 40 ng/ml, Sigma) until day 37. KOSR-free media was
which demonstrate the differentiation of retinal cell types from then supplemented with rmNoggin (10 ng/ml), rhDkk1 (10 ng/
pluripotent cells, and by mimicking the chemical and cytokine ml), rhIGF-1 (10 ng/ml), rhbFGF (bFGF, 5 ng/ml), retinoic acid
microenvironmental signals known to guide retinal histogenesis (500 nM, Sigma), T3 (40 ng/ml), taurine (2 lm, Sigma), and Shh
during normal development, we have developed an adapted (12 nM) until day 60. During days 37-41 (Fig. 1A, black dashed
differentiation protocol to promote photoreceptor differentiation line), PR-induction medium was supplemented with Human
from hESC and hiPSC cultures. This method combines some Activin-A (100 ng/ml, Peprotech), to encourage the exit of pho-
aspects of previously published protocols on hESC and toreceptor progenitor cells from the cell cycle and their further
hiPSC differentiation toward the retinal lineage [8, 12], which maturation [25]. This induction method combines growth factors
we have then further modified with additional supplements that at defined concentrations shown to induce photoreceptor differ-
were added on the basis of reports from retinal and eye field entiation from two previous studies [8, 12] plus our own addition
developmental papers. We show that our serum and feeder-free of Activin A, Shh (known to cause an increase in the number of
differentiation method results in the generation of photoreceptor retinal progenitors and photoreceptor cells) and T3 (known to
progenitors and RPE from hESC and hiPSC with higher effi- promote cone photoreceptor differentiation in human fetal retinal
ciency than what has previously been achieved. Our data also cultures; [26]). Two different concentrations were used for Shh;
suggest that at least some hESC and hiPSC lines are endowed 30 nM during the initial stages of differentiation (previously
with an endogenous potential to generate neural and retinal cells defined as optimal for inducing ventral forebrain specification;
under minimal culture conditions that can be enhanced and/or [27, 28]) and 12 nM in the last stage of differentiation (shown to
accelerated by the addition of specific mitogens and metabolites promote differentiation of embryonic rat retinal cultures toward
(Dkk1, B27, and Noggin) during the initial phase of differentiation. a photoreceptor phenotype; [29]). Minimal control cultures
[con()] consisted of hESC/hiPSC population that were differen-
tiated in basal media alone (Fig. 1A, green dashed line), in the
absence of B27/N2/additional factors. For more information,
MATERIALS AND METHODS refer to Supporting Information.
hESC and hiPSC Culture and Differentiation Analyses
hESC (line H9, WiCell, passage 34-45) and hiPSC generated Quantitative Reverse Transcription-Polymerase Chain
from human neonatal dermal skin fibroblasts (hiPSC-NHDF, Reactions. Total RNA was extracted using TRIzol reagent
Mellough, Sernagor, Moreno-Gimeno et al. 675

Downloaded from https://academic.oup.com/stmcls/article/30/4/673/6415666 by CINVESTAV user on 26 May 2022

Figure 1. Three-step induction protocol and examples of the resulting culture morphology. (A): Schematic showing the different stages of the
60-day differentiation protocol. Colored broken lines represent the different stages of media supplementation (Materials and Methods). (B–J):
The morphology of human embryonic stem cells differentiating under (B–D, I, J) control and (E–G, H) photoreceptor-induction conditions at (B,
E) 35 days, (C, F) 45 days, and (D, G, H) 60 days of differentiation. (H): Cells differentiating under conditions designed to induce photoreceptor
production generated neural-like populations with a proportion of cells exhibiting long neurites. (I, J): The typical appearance of retinal pig-
mented epithelial-like cells in control cultures on day 35 of differentiation under (I) phase microscopy and (J) following confocal microscopy
counterstained with Hoechst 33342. KOSR, knockout serum replacement. Scale bars: B, C, E, F ¼ 200 lm, D, G, H ¼ 100 lm, and I, J ¼ 50
lm. Abbreviation: PR, photoreceptor.

(Invitrogen, Paisley, UK) according to manufacturer’s instructions. tions were carried out and samples analyzed using an AB7900HT
Following DNase treatment using RQ1 DNaseI (Promega, South- real-time analyzer. All primers used in this study are listed in the
ampton, UK), cDNA was synthesized using SuperScript III Supporting Information Table S2. Data were analyzed using qBase
Reverse Transcriptase (Invitrogen). Quantitative reverse transcrip- v1.3.5 and the comparative threshold cycle (Ct) method. In all
tion-polymerase chain reactions (qRT-PCR) analysis was carried samples, the results were normalized to the geometric mean of
out using SYBR GreenER PCR master mix (Invitrogen). All reac- three housekeeping genes GAPDH, SDHA, and RPL13A, and
www.StemCells.com
676 Efficient Photoreceptor Generation from Human Stem Cells

referenced to hESC, 7-day differentiated hESC, human retina or libur system (BD Biosciences) with CellQuest Pro software
human RPE in accordance with the gene of interest. (BD Biosciences). At least 10,000 events were analyzed in
each experiment.
Array-Comparative Genomic Hybridization. To ensure that Statistical Analysis
the hESC line (H9) used in this study had not acquired any
chromosomal abnormalities during ex vivo expansion, we per- Two-tailed paired Student’s t test was used to analyze results
formed array-comparative genomic hybridization (aCGH) as between groups at various time points during the differentia-
outlined in Supporting Information. The analysis did not show tion process and between control and PR-induced groups at
significant abnormalities (Supporting Information Fig. S1A– the same stage of differentiation. Results were considered
S1C), with the exception of two close amplifications of 186 significant if p < 0.05.
and 458 kb at 14q23.2 and 14q23.3, which correspond to
known copy number variations (Database of Genomic Var-

Downloaded from https://academic.oup.com/stmcls/article/30/4/673/6415666 by CINVESTAV user on 26 May 2022


iants at http://projects.tcag.ca/variation/). We also noticed a RESULTS
deletion of 2MB at 6p22.2-p22.1; however, in-depth analysis
suggested that the deletion observed at chromosome 6 and
To design a more efficient photoreceptor differentiation proto-
amplifications observed at chromosome 14 were not signifi-
col for pluripotent cell types, we have combined some aspects
cant as they did not meet the criteria of significant genomic
of previously published protocols on hESC and hiPSC differ-
aberrations, which is the presence of three consecutive probe
entiation toward a retinal lineage [8, 12] and then made addi-
alterations (at chromosome 6p22.2-p22.1, three nonconsecu-
tional modifications on the basis of reports from retinal and
tive deleted probes were found in a region comprising 77
eye field developmental papers. Akin to previously published
probes [Supporting Information Fig. S1B], while at 14q23.2
methods [8, 12], our differentiation protocol (Fig. 1A) was
and 14q23.3, only two consecutive probes were amplified,
designed to guide the transition of cells from a pluripotent
respectively [Supporting Information Fig. S1C]). We also
state to that of the retinal lineage, more specifically, a photo-
investigated the copy numbers of 61 reported stem cell-associ-
receptor fate by sequentially exposing cells to antagonists and
ated genes [30] and this analysis showed no significant differ-
cytokines known to direct: (a) a ventral neural fate; (b) eye
ences in DNA copy number over the entire chromosomal
field/retinal determination; and (c) photoreceptor specification
regions. In summary, our aCGH analysis indicated that H9
and maturation. hESC/hiPSC were differentiated under this
hESC used in this study did not show genomic instabilities
defined culture protocol (named PR-induction, red line in
that could impact upon their pluripotency.
Fig. 1A) for a period of 60 days and analyzed by qRT-PCR,
flow cytometry, and immunocytochemistry. Control cultures
Fluorescence Immunocytochemistry and Microscopy. Cells were set up in parallel in the absence of growth factors and/or
were fixed in 4% (w/v) paraformaldehyde for 15 minutes. culture supplements and named appropriately in each figure
Permeabilization (0.2% (v/v) Triton X-100 in PBS) was (blue and green lines in Fig. 1A).
performed prior to staining with antibodies for internal cell
markers. A blocking step was performed by incubation in The Loss of Pluripotency and Acquisition of
antibody diluent (1% (w/v) bovine serum albumin, Sigma, a Neural Lineage
with 5% (v/v) normal goat serum, Invitrogen) prior to staining Differentiation of hESC under control and PR-induction condi-
for 30 minutes. Cells were incubated with anti-rhodopsin tions yielded no discernable differences in culture morphology
(Sigma), anti-opsin blue (OPN1LW, Millipore), anti-opsin at the EB stage. qRT-PCR analysis of gene expression demon-
red/green (OPN1SW, Millipore, Watford, UK), anti-RPE65 strated the rapid downregulation of pluripotency markers
(AbCam, Cambridge, UK) antiprimary antibodies for 1 hour NANOG and OCT4 over the first 30 days, indicating the loss
and secondary antibodies for 30-60 minutes. Nuclei were of pluripotency (Fig. 2). Following transfer to adherent culture
counterstained with 10 lg/ml Hoechst 33342 then cultures conditions, EBs attached to generate proliferative cultures (Fig.
coverslipped using Vectashield (Vector Laboratories, Peter- 1B–1H) with many cells displaying neural-like morphology.
borough, UK). Bright-field and fluorescent images were Neural cells exhibit a variety of morphology depending on
obtained using a Zeiss inverted microscope and AxioVision their phenotype and stage of maturation. Neural progenitor
software. Immunopositive cells were quantified manually by cells are often bipolar in shape with a small soma and shorter,
randomly sampling and photographing 10 fields of view at less developed neurites than their mature neuronal and glial
20 magnification. All Hoechst-labeled nuclei and immuno- counterparts (e.g., Fig. 1D). Late stage neurons can differ
positive cells within these confines were counted. vastly in their soma size as well as their polarity (ranging from
unipolar to multipolar), dendritic arborization and axon length,
Flow Cytometric Analysis. Differentiating cells were disso- some cells exhibiting axons which traverse great distances
ciated by treatment with Accutase (Gibco) and then processed (e.g., as in Fig. 1D–1H). Pigmented foci, indicating the emer-
in 35 lm Medicons using a Medimachine (Becton Dickinson, gence of RPE cells, appeared within floating EB cultures over
New Jersey). Cells were collected by centrifugation at 250g days 20-30 and following transfer to adherent culture condi-
for 5 minutes and fixed first in 0.5% paraformaldehyde at tions generated sheets of pigmented epithelial cells resembling
37 C for 10 minutes, then by treatment with 90% methanol native RPE (Fig. 1I and 1J). This occurred with higher fre-
for 30 minutes on ice. Cells were washed in PBS then trans- quency in control cultures, possibly due to the addition of
ferred to incubation buffer (0.2% BSA and 5% goat serum in bFGF in PR cultures, which has been shown to suppress RPE
PBS) for 10 minutes at room temperature followed with un- induction [10, 13].
conjugated anti-Chx10, anti-CRX, anti-rhodopsin, anti- qRT-PCR analysis of PR-induced cultures revealed low
OPN1LW, anti-OPN1SW, or anti-RPE65 primary antibody levels of trophoctoderm (CDX2), endoderm (GATA4, AFP),
for 1 hour before incubation with fluorescein (FITC) second- and mesoderm (BRACHYURY) markers after day 30 of differ-
ary antibody (1:800, Sigma) for 30 minutes at room tempera- entiation and an upregulation of the primitive ectodermal-
ture. Samples were washed in PBS and protected from light associated gene FGF5 (Fig. 2, red bars). This occurred
prior to analysis. Analysis was performed using the FACSCa- alongside an increase in SOX2 and NCAM1 gene expression
Mellough, Sernagor, Moreno-Gimeno et al. 677

Downloaded from https://academic.oup.com/stmcls/article/30/4/673/6415666 by CINVESTAV user on 26 May 2022


Figure 2. Pluripotency and germline gene expression in differentiating human embryonic stem cells (hESC) over 60 days. Assessment of
pluripotency markers NANOG and OCT4 and lineage markers from all three germ layers in differentiating control (blue bars) and PR-induced
population (red bars) over 60 days by quantitative reverse transcription-polymerase chain reactions. All data (Y-axis) are presented as the normal-
ized ratio of gene expression to housekeeping genes GAPDH, SDHA, and RPL13A. NANOG and OCT4 are calibrated to hESC (hESC ¼ 1), all
other genes to hESC that had been differentiated for 7 days (H9d7 ¼ 1). Data ¼ mean 6 SEM, n ¼ 3. Abbreviations: FGF, fibroblast growth
factor; PR, photoreceptor.

over the differentiation period (Fig. 4A), indicating the emer- period while the early retinal marker PAX6 increased in PR-
gence of neuroectodermal derivatives. In control cultures induced cultures over time (p ¼ .048) from day 45 to reach a
however, some expression of AFP, CDX2, and GATA4 was prominent peak at day 60 (Fig. 4A). Flow cytometric analysis
maintained over the 60 days of differentiation (Fig. 2, blue detected the greatest number of cells expressing the retinal
bars), with observed levels of FGF5 expression lower than in progenitor marker CHX10 on day 15 of differentiation in the
mitogen-treated population, indicating spontaneous differentia- PR-induced group (Fig. 4B, red line) showing an almost two-
tion toward other germ layers. fold increase over control population at this time (expressed
in 25% of control population and 43% of PR-induced). After
The Emergence of a Retinal Lineage day 30, both population follow a similar expression course
We went on to assess the emergence of eye field and retinal and decline from 13% of cells at day 45 (Fig. 3A) to only a
progenitor markers in cultures differentiating under our fragment of the population expressing this antigen by day 60
PR-induction and control regimens (Figs. 3, 4). NESTIN (<1%, Fig. 4B). No significant changes were, however,
expression remained constant throughout the differentiation detected in CHX10 transcript levels during the differentiation
www.StemCells.com
678 Efficient Photoreceptor Generation from Human Stem Cells

Downloaded from https://academic.oup.com/stmcls/article/30/4/673/6415666 by CINVESTAV user on 26 May 2022


Figure 3. The expression of retinal and photoreceptor-specific markers in differentiating human embryonic stem cells by immunofluorescence
histochemistry and flow cytometry. (A): Flow cytometric analysis of CHX10, CRX, Opsin blue (OPN1SW), Opsin red/green (OPN1LW), and
RHODOPSIN under control (con) and photoreceptor-induction conditions (PR) at 45 and 60 days. (B–D): Immunofluorescent staining of (B, C)
RHODOPSIN (green) and ZO-1 (red) counterstained with Hoechst (blue) and (D) cone-specific OPN1SW (red) near cells immunopositive for the
RPE marker RPE65 (green) at 45 days. Scale bars ¼ 50 lm. (E): CRX, OPN1SW, and RHODOPSIN expression as detected by flow cytometry
in H9 cells differentiated under control (con) and PR-induction conditions at 45 days. Values equal mean 6 SEM (n ¼ 3). Both cultures demon-
strated expression of photoreceptor-specific markers at this time point. Abbreviation: RPE, retinal pigmented epithelium.

period and also between control and PR-induced groups. This revealed an early peak in RPE65 expression at day 30 in the
might be explained by differences between quantification PR-induced group (Fig. 4A, 4B). In control cultures, we
methods, the sensitivity of each method and the measurable observed the emergence of RPE cells over the first 30 days
outcome (protein vs. transcript) with flow cytometric analysis which, unlike PR-induced cultures, continued to rise until day
assessing the percentage of CHX10 immunopositive cells 60 (Fig. 4A, RPE65, SILV) consistent with published litera-
(and not total level of CHX10 protein expression) and qRT- ture, which indicates spontaneous emergence of these cells in
PCR measuring the total level of the CHX10 transcript. the absence of exogenous growth factors [10, 13, 15]. The
RPE65 is a cytoplasmic protein involved in retinoid me- expression of eye field and retinal progenitor markers PAX6,
tabolism [31, 32]. Flow cytometric and qRT-PCR analysis CHX10, and RPE-specific RPE65 alongside the appearance of
Mellough, Sernagor, Moreno-Gimeno et al. 679

Downloaded from https://academic.oup.com/stmcls/article/30/4/673/6415666 by CINVESTAV user on 26 May 2022

Figure 4. The expression of differentiation-associated genes and antigens. (A): Quantitative reverse transcription-polymerase chain reaction analy-
sis of neuroectodermal (SOX2, NCAM1, NESTIN, and NEUROD), retinal progenitor (PAX6 and CHX10), photoreceptor progenitor (CRX), phototrans-
duction pathway (RECOVERIN, PDE6b, OPSIN, and ARR3), and retinal pigmented epithelial (RPE65 and SILV) gene expression in control (blue
bars) and mitogen-treated population (PR, red bars) over the 60 days of differentiation. Data (Y-axis) is normalized to housekeeping genes and
referenced to human embryonic stem cells (hESC; SOX2, NCAM1, NEUROD, NESTIN, and PAX6, hESC ¼ 1), human retina (CHX10, CRX, RECOV-
ERIN, PDE6b, OPSIN, and ARR3, human retina ¼ 100), or human RPE (RPE65 and SILV, RPE ¼ 1). Values equal mean 6 SEM (n ¼ 3). (B): Flow
cytometric analysis of PR-induced (red line) and control cultures differentiated with B27/N2 (con, blue line) against control() cultures differentiated
in the absence of B27/N2 (con(), green line) revealed reduced immunopositivity for CHX10, RPE65, and OPN1SW in con() cultures and an
important role for B27/N2 in the levels of retinal-specific gene expression previously observed in control population (A, blue bars) in the absence of
any other mitogens or antagonists. Values equal mean 6 SEM (n ¼ 3). Abbreviations: PR, photoreceptor; RPE, retinal pigmented epithelium.

www.StemCells.com
680 Efficient Photoreceptor Generation from Human Stem Cells

pigmented foci over the first 30 days of differentiation indi- available KOSR [33]; therefore, it is not likely to be the pres-
cates the emergence of early retinal-specific phenotypes from ence of KOSR during the first 37 days of differentiation
differentiating hESC. which enabled these effects. Some constituents of the neural
supplement B27 are demonstrated to promote neuralization,
The Emergence of a Photoreceptor Phenotype enhancing neural progenitor proliferation and differentiation
Following the transfer of differentiating hESC to adherent cul- as well as the development of the machinery necessary for
ture conditions, we assessed the expression of photoreceptor phototransduction [34–36]. This suggests that B27, a compo-
markers by qRT-PCR, flow cytometry and immunofluorescent nent of our basal ventral neural induction medium and there-
histochemistry (Figs. 3, 4). Flow cytometric analysis on day fore present in control cultures during differentiation, may
45 of differentiation confirmed the persistence of some play a role in the specific differentiation of hESC towards ret-
CHX10þ cells (up to 13%) and a similar number of cells inal cells. We therefore questioned whether the removal of
were immunopositive for the early postmitotic photoreceptor this supplement over the first 37 days of differentiation fol-

Downloaded from https://academic.oup.com/stmcls/article/30/4/673/6415666 by CINVESTAV user on 26 May 2022


precursor marker CRX (up to 16%, Fig. 3A). Over days 45- lowed by the combined removal of B27 and N2 in the final
60, a marked increase was observed in CRX gene expression window of differentiation (days 37-60) would affect differen-
under control induction conditions (Fig. 4A). This led us to tiation potential.
test whether any differentiating cells within these populations To investigate this, we introduced an additional control
might be expressing other photoreceptor markers. This into our experiments, by differentiating hESC cultures in the
included the neuronal calcium-binding protein RECOVERIN, absence of B27 or N2 in a minimal media [named control()
ARRESTIN 3 which is a marker of outer segments of red, cultures, refer to Figure 1A green line]. hESC differentiating
green, and blue cone photoreceptors, and cone-specific light- under these conditions lost their pluripotency marker expres-
sensitive G-protein-coupled receptors, or opsins. Indeed, sion in a similar way to control and PR-induced groups (Sup-
from days 45 to 60 of differentiation, an increase in the porting Information Fig. S2). The removal of B27, followed
expression of ARRESTIN 3 was detected under control condi- by combined B27 and N2 removal from the differentiation
tions (Fig. 4A). media resulted in a reduction in early expression levels of the
Flow cytometric analysis revealed the highest expression retinal progenitor marker CHX10 when compared with sup-
of photoreceptor markers RHODOPSIN, cone-specific opsin plemented cultures; but this was, however, followed with by a
blue (OPN1SW) and opsin red/green (OPN1LW) at day 45 of steady increase in CHX10 over days 15-60 (Fig. 4B), reach-
differentiation (found in 18%, 52%, and 60% of cells cultured ing by day 60 the same expression level as observed in B27/
under PR-inducing conditions, respectively, Fig. 3A, 3E). By N2 supplemented cultures at day 15 of differentiation. Similar
day 60, this proportion had declined, resulting in almost com- results were also observed for the RPE marker RPE65, where
plete loss of RHODOPSIN (1.1%) and only a small number the same expression levels were observed for PR-induced,
of OPN1SW ( 4%) immunopositive cells remaining, while control and control() groups on day 60 of differentiation
13% of the population remained OPN1LWþ at this time (Fig. 4B). Despite this, hESC differentiating under control()
(Fig. 3A). The expression of photoreceptor-specific markers conditions failed to reach the peak OPN1SW expression
was confirmed following immunocytochemical analyses and observed at day 45 in PR-induced and control cultures. Both
manual cell counts (Fig. 3B–3D). sets of results shown in Figures 3E and 4B suggest no signifi-
Detection of the postmitotic photoreceptor marker CRX cant differences in early and mature photoreceptor marker
alongside late-stage photoreceptor-specific markers indicates expression between control and PR-induced populations.
the emergence of hESC-derived photoreceptors within cul- Given that B27 (present throughout differentiation) and N2
tures differentiated using our protocol. Comparison of control (added in the final window of differentiation after day 37,
and PR-induced cultures by qRT-PCR, however showed only refer to Fig. 1A) are the only common culture components
one statistically significant increase in the expression between PR-induced and control groups and are both missing
of OPSIN in PR-induced population observed at day 60 (p ¼ from the control() group, this suggests a role for these two
.035, Fig. 4A). In addition, flow cytometric analysis from day components in enhancing and accelerating the endogenous
30 onward showed no significant changes in the expression of capacity of hESC cultured under minimal culture conditions
photoreceptor-specific markers (Figs. 3E, 4B) and, in most to undergo neural and retinal differentiation.
cases, control and PR-induced populations followed a similar Similar results were obtained during the differentiation of
time course with regard to the onset of neuroectodermal, reti- hiPSC-NHDF cells (Supporting Information Fig. S3). Expres-
nal progenitor and photoreceptor-specific markers and levels sion of both cone-specific opsin blue (OPN1SW) and opsin
of gene expression, suggesting that perhaps an endogenous red/green (OPN1LW) at day 45 of differentiation was similar
and compensatory growth factor signaling pathway is active across control and PR-induced cultures, but lower in con-
in control cultures, corroborating findings reported by Meyer trol() cultures, while the opposite was observed for early
et al. [11]. retinal marker, PAX6 (Supporting Information Fig. S3).
Together, these results indicate an endogenous, although
somewhat delayed, capability of differentiating hESC cultures
Further Investigation into the Conditions Enabling to produce certain neural and retinal phenotypes during
Retinal Specification from Differentiating hESC prolonged culture in the absence of specific inductive cues.
Reveals an Important Role for B27 and N2
As shown above, we observed that control populations differ-
entiated under basal media conditions were capable of exhib- The Endogenous Expression of Key Signaling
iting neural, eye field, retinal and photoreceptor gene expres- Factors in hESC
sion over time. This was an unexpected result and we set out The endogenous expression of the Wnt antagonist DKK1 and
to address the role of specific media components during FGF signaling has previously been reported in hESC differen-
hESC differentiation. Human ESCs express the receptors for tiating toward the eye field [11]. Endogenous SHH signaling
fibroblast growth factor (FGF), activin A, epidermal growth in mouse ESC (mESC) can promote a ventral neural fate [37]
factor (EGF), nerve growth factor (NGF) and retinoic acid; and, although endogenous SHH is very low in hESCs, levels
however, the ligands for these are all absent in commercially of SHH become enriched upon EB differentiation [38]. We
Mellough, Sernagor, Moreno-Gimeno et al. 681

Downloaded from https://academic.oup.com/stmcls/article/30/4/673/6415666 by CINVESTAV user on 26 May 2022


Figure 5. Assessment of endogenous growth factor and antagonist gene expression. (A): Quantitative reverse transcription-polymerase chain
reactions (qRT-PCR) analysis of EGF, NGF, SHH, NODAL, and DKK1 expression was performed at 15-day intervals over the differentiation
period in control (with B27/N2 only, blue line), minimal control (no B27/N2/mitogen supplementation, green line) and in B27/N2/mitogen-
supplemented PR-induced population (PR, red line). (B): qRT-PCR analysis showing delayed onset of NEUROD expression in control()
cultures compared with control and PR-induced population. (C): qRT-PCR analysis revealed that, of all conditions tested, the highest expression
of CRX and RECOVERIN on day 60 was seen in control and PR-induced cultures. (A–C): Data (Y-axis) are normalized to housekeeping genes
and calibrated to human embryonic stem cells ¼ 1 for panel A and human retina for panels B and C (human retina ¼ 1). Values equal mean 6
SEM (n ¼ 3). Abbreviations: EGF, epidermal growth factor; NGF, nerve growth factor; PR, photoreceptor; and SHH, human sonic hedgehog.

therefore investigated the expression of various factors in expression of EGF, NGF, NODAL, and SHH was observed
hESCs differentiating under our protocol. qRT-PCR results from day 45 to day 60 in these cultures, reaching levels that
indicated that endogenous levels of EGF, NGF, and SHH rose were greatly elevated in comparison with control and PR-
in hESC cultures over the differentiation period, NODAL was induced population (Fig. 5A). DKK1 levels were also higher
strongly expressed on day 15 but then rapidly declined, while in control() cultures suggesting elevated endogenous Dkk1
expression of DKK1 remained relatively constant after this signaling in hESC differentiating under minimal culture con-
time point. We observed no significant difference in the en- ditions and corroborating previously published data [11].
dogenous expression of EGF, NGF, SHH, NODAL, or DKK1 We then went on to assess the effects of each of our three
between control and PR-induced cultures (Fig. 5A, blue and culture conditions; control(), control, and PR-induced on the
red lines). expression of neuroectodermal, retinal, and photoreceptor
When these results were compared with control() popu- genes. Our analysis demonstrated that hESC differentiating
lation, some differences emerged (Fig. 5A, green lines). Akin for 60 days without B27 and N2 resulted in a delay in the
to our observations of the delayed upregulation of early reti- onset of neuroectodermal gene expression when compared
nal markers in control() cultures, increased endogenous with normal controls and PR-induced population (Fig. 5B),
www.StemCells.com
682 Efficient Photoreceptor Generation from Human Stem Cells

Downloaded from https://academic.oup.com/stmcls/article/30/4/673/6415666 by CINVESTAV user on 26 May 2022


Figure 6. Further analysis of the individual contribution of B27/N2 supplements and added factors on endogenous gene expression and embry-
oid body morphology. (A): Morphological differences were observed between EBs from (a) control() and (b) control cultures as well as during
supplementation with individual factors. T3 treatment promoted the formation of small, highly pigmented EBs (c, d), LeftyA induced the forma-
tion of polarized EBs with a prominent axis (e), while in IGF-1-treated population disc-shaped EBs emerged which contained numerous embed-
ded optic cup-like structures (f, g) only rarely seen to contain pigmented cells (g). (B): PAX6 and CHX10 expression at days 15 and 30 was
enhanced in cultures treated with Dkk1, noggin, or T3 during the first 30 days of differentiation. Data (Y-axis) are normalized to housekeeping
genes. Results for PAX6 and CHX10 are calibrated to human retina (human retina ¼ 1). Values equal mean 6 SEM (n ¼ 3). Abbreviations: IGF,
insulin-like growth factor; shh, human sonic hedgehog; and T3, 3,30 ,5-triiodo-L-thyronine.

consolidating the flow cytometry data analysis shown in Further Analysis of the Individual Contribution of
Figure 4B. Nonetheless, by qRT-PCR analysis, the cultures Each Factor During hESC Differentiation
demonstrating the best CRX and RECOVERIN expression on
day 60 across all groups were the control (with B27/N2) and To test the individual contribution of each component of the
PR-induced populations (Fig. 5C) corresponding with the data PR-induction media, we supplemented differentiation media
shown in Figure 4B and once more suggesting that at least with isolated factors during floating EB culture for up to 30
B27 and/or N2 (present in both control and PR-induced days. There were some apparent differences in gross EB
cultures) are two important media components that enhance morphology between normal controls (with B27) and mini-
the induction of photoreceptor-specific gene expression in dif- mal controls (no B27) at this stage of differentiation
ferentiating hESC. (Fig. 6A; panels a and b, respectively) with more cystic EBs
This suggests that, in the absence of external cues, a pro- appearing in the minimal control group. In addition, some
portion of hESCs follow an intrinsic neural and retinal differ- interesting morphological differences emerged following
entiation pathway, which is associated with the delayed onset treatment with individual factors. The addition of T3 pro-
of neural and retinal markers under minimal culture condi- moted the formation of small, highly pigmented EBs
tions. This capability, as seen in minimal controls, may be (Fig. 6A, panels c and d), while LeftyA induced the forma-
due to the upregulation of endogenous signaling factors start- tion of polarized EBs containing a prominent axis (Fig. 6A,
ing as early as the first 15 days (DKK1) but from our observa- panel e). Interestingly, differentiation with IGF-1, shown
tions in most cases between days 30 and 60 (EGF, NGF, specifically to induce eye formation in Xenopus and an
NODAL, and SHH). Nonetheless, the induction of enhancer of retinal progenitor gene expression [39], resulted
photoreceptor marker expression remained more efficient in in the appearance of disc-shaped EBs containing numerous
control and PR-induced cultures. visible internal neural rosette-like structures (Fig. 6A, panels
Mellough, Sernagor, Moreno-Gimeno et al. 683

Downloaded from https://academic.oup.com/stmcls/article/30/4/673/6415666 by CINVESTAV user on 26 May 2022

Figure 7. Further analysis of the directed differentiation of human embryonic stem cells (hESC) with different combinations of supplements and
mitogens revealed variance in the expression of mitogens, mature retinal genes, and culture morphology. Treatment of hESC with B27/N2 and all fac-
tors for 37 days and then with isolated factors for the remainder of the differentiation period induced variation in (A) endogenous gene expression and
(B) photoreceptor and RPE-specific genes on day 60 of differentiation. All treatments generally yielded heterogeneous neural-like cultures (C–I) with
the presence of RPE sheets displaying distinct boundaries (D, E). Cells within adherent EBs commonly generated long processes, some of which
extended hundreds of microns (C). Optic-cup-like structures that were observed following IGF-1 treatment during floating culture were maintained
following transfer to adherent conditions in PR-induction media (F). These were also observed in population differentiated with PR-induction media
over the first 37 days and then with either T3 (G) or taurine (H), and in lower frequency in cultures grown under PR-induced conditions for the full dif-
ferentiation period (I). Data are normalized to housekeeping genes and referenced to hESC (panel A, hESC ¼ 1), human retina (panel B, human retina
¼ 1), or human RPE (RPE ¼ 1, RPE65, panel B). Values equal mean 6 SEM (n ¼ 3). Scale bars: C–E, G, I ¼ 100 lm and F, H ¼ 400 lm. Abbrevia-
tions: bFGF, basic fibroblast growth factor; EGF, epidermal growth factor; FGF, fibroblast growth factor; IGF-1, insulin-like growth factor 1; NGF,
nerve growth factor; PR, photoreceptor; RPE, retinal pigmented epithelium; SHH, human sonic hedgehog; and T3, 3,30 ,5-triiodo-L-thyronine.

www.StemCells.com
684 Efficient Photoreceptor Generation from Human Stem Cells

f and g), which have also been reported under minimal cul- early phase of differentiation, while the majority of
ture conditions [16]. CRXþphotoreceptor progenitors were generated over days 30-
Our quantitative RT-PCR expression analysis detected the 45. The onset of mature photoreceptor markers at this time
best CHX10 expression in cultures exposed to B27 plus Dkk1 indicated that the emergence of the two distinct photoreceptor
or B27 plus noggin (Fig. 6B). In cultures supplemented with types, rods and cones, was also concordant with what one
B27 and one other factor only, PAX6 expression was also might expect developmentally; cone photoreceptors are born
found to be higher than PR cultures differentiated with all during the early phase of retinal histogenesis while rods
factors together (Fig. 6B). Endogenous Dkk1 signaling has emerge later [44]. Our gene expression results support this
been implicated in acquisition of early retinal markers (PAX6 observation, showing elevated levels of cone-specific OPSIN
and RAX) during differentiation of hESC [11]. To investigate on day 30, prior to the peak of rod-specific gene expression
this further in our differentiation protocol, we performed flow on day 60.
cytometric analysis in Dkk1-supplemented control cultures We also report an endogenous capacity of hESCs and

Downloaded from https://academic.oup.com/stmcls/article/30/4/673/6415666 by CINVESTAV user on 26 May 2022


(but devoid of all other added factors) and observed a 76.6% hiPSC to generate neural and retinal phenotypes under
increase in number of CRX expressing cells and a 7.8% minimal culture conditions. This capability has previously
decrease in number of cone-specific opsin blue (OPN1SW) been demonstrated in hESC and hiPSC [11, 16], but is not a
expressing cells (data not shown), suggesting an important widely published phenomenon and it may be due to the en-
role for Dkk1 in enhancing the specification of photoreceptor dogenous production of various signaling molecules and
precursors but not later stage photoreceptor cells. growth factors from the heterogeneous mix of cells present in
In cultures exposed to PR-induction conditions for the first embryoid body cultures generated from pluripotent stem cells
37 days then swapped to minimal media either alone or as demonstrated by this study and others. Trophic support for
supplemented with isolated factors, we observed that some emerging retinal cell types from neighboring RPE cells is
factors elicited similar results (Fig. 7A). For example, hESC another potential explanation for the elevation in retinal-spe-
differentiated under minimal conditions (no B27/N2/factors) cific markers observed in populations differentiated in the ab-
from days 37 to 60, or in minimal media supplemented with sence of mitogens or supplements [45].
Dkk1, N2, or T3 resulted in the elevated endogenous expres- A recent groundbreaking paper demonstrates that struc-
sion of FGF2, SHH, DKK1, EGF, and NGF (Fig. 7A). We tures reminiscent of the optic vesicle can arise from mESCs
went on to assess the effect of each media component on the differentiated in low-concentration matrigel and minimal dif-
expression of retinal and photoreceptor genes. Of mitogen- ferentiation medium [46], highlighting a propensity for mESC
treated groups, our analysis demonstrated the best expression to undergo stepwise differentiation and self-organization
of OPSIN, PDE6b, and ARRESTIN3 on day 60 in cultures under very simple culture conditions. This corroborates and
that had been differentiated under PR-induction conditions for extends previously published data showing that mouse ESC
37 days and then either with no factors or supplemented with cultured under minimal conditions and without instructive sig-
N2, Dkk1, or T3 (Fig. 7B). These qRT-PCR results were con- naling can undergo neural differentiation [47]. We also
firmed by flow cytometry data, which showed an increase in observed the emergence of neural rosette-like structures from
the number of CRXþ (84%), OPN1SWþ (89.2%), and hESCs and hiPSC during the first 30 days of differentiation.
OPN1LWþ (34.6%) cells in cultures that had been differenti- These were most commonly observed during the early stages
ated under PR-induction conditions for 37 days and subse- of differentiation in cultures treated with IGF-1, a known in-
quently under minimal conditions compared to those ducer of eye formation and a potent enhancer of retinal pro-
subjected to PR-inducing conditions for the entire differentia- genitor gene expression [8, 39]. During the latter stages of
tion period (data not shown). Together these data suggest an differentiation, we also observed numerous rosette-like struc-
important role for B27, Dkk1, and Noggin in the first 37 days tures in populations that had been treated with all supplements
of differentiation. In addition, they confirm that although the and mitogens for photoreceptor-induction over the first 37
addition of N2, Dkk1, or T3 during the final stage of differen- days, followed by treatment with T3 or taurine only. Some
tiation can facilitate the yield of the mature photoreceptors, were also observed in cultures differentiated under our photo-
this is not a requirement for further cell maturation to occur. receptor-induction regime for the entire differentiation period,
albeit with less frequency. These rosette-like structures are
similar to optic vesicle-like structures reported by Meyer
et al. [16] during hESC/hiPSC differentiation, and current
DISCUSSION work within our group is focusing on identification of the
RPC types present therein and their potential to generate func-
The ability to generate expandable populations that can yield tionally mature photoreceptor and RPE cells.
a large number of photoreceptors is a prerequisite for the suc- One previous study has shown that the treatment of
cessful application of cell replacement therapy for outer reti- hESCs with B27, Dkk1, noggin, IGF-1 and subsequently with
nal degeneration. Our strategy for photoreceptor production N2 and FGF can result in more than 80% of cells expressing
from hESC and hiPSC relies upon the use of various factors retinal progenitor markers [8]. However, although 12% of the
and antagonists that are known to play a role in neural induc- population expressed CRX, less than 0.01% were found to be
tion and retinal histogenesis during development and from in positive for mature photoreceptor markers. Osakada et al.
vitro studies [8, 11, 12, 21, 25, 40-42]. Our aim was to de- achieved CRX expression in 11% of the population following
velop a protocol to increase the yield of photoreceptors from 120 days of differentiation, which increased to around 20%
hESC and hiPSC in a shorter time frame than what has been by 170 days, with 5% of cells expressing Rhodopsin by day
achieved previously. 150 [12]. Meyer et al. achieved 55.9% of CRX expressing
Our results indicate that our protocol directs the stage- cells (at day 80 of differentiation), which undergo differentia-
specific differentiation of hESC and hiPSC toward a neural tion to photoreceptor-like cells displaying characteristic
fate, followed by retinal field and then photoreceptor lineage. electrophysiological responses [16]. Each of these studies rep-
The appearance of these phenotypes occurred in a sequential resents great achievement (Supporting Information Table S1
manner consistent with retinal development in vivo [43]. We for more detail). Yet for this therapy to be translated to the
observed that CHX10þ retinal progenitors emerged during the clinic, it will be advantageous if this process was more
Mellough, Sernagor, Moreno-Gimeno et al. 685

efficient. We set out to try and improve existing protocols and of differentiation, the further maturation of these cells to
from our results we report that we are able to generate 16% express mature photoreceptor markers was most efficient in
of CRXþ cells and 52% of cone-like photoreceptor cells the absence of added supplements during the final stage of
within 45 days of differentiation. Although this differentiation differentiation. These findings are in close resonance with the
period is longer than what has been reported in one earlier reported propensity of mESC to undergo stepwise differentia-
study [8], the higher efficiency of generated cells that express tion resulting in the formation of self-organized optic cup
mature photoreceptor markers compared to previous studies structures under very simple culture conditions [46] and while
after 45 days of differentiation (refer to Supporting Informa- it remains to be investigated whether human pluripotent stem
tion Table S1) strongly suggests that our three-step differen- cells are endowed with such intrinsic self-organizing capacity,
tiation protocol extends and improves current achievements in the data presented in this manuscript on the role of various
the hESC/hiPSC differentiation field. Notwithstanding the metabolites and growth factors provide a solid platform for
increased efficiency, our protocol has also its limitations. We initiating such investigations.

Downloaded from https://academic.oup.com/stmcls/article/30/4/673/6415666 by CINVESTAV user on 26 May 2022


observed the rapid loss of cells committed to a photoreceptor Endogenous growth factor signaling specific to each
fate (CRXþ/OPSINþ/RHODOPSINþ) over days 45-60. This human pluripotent cell line has been shown to affect the dif-
is similar to observations in a study using cultured mouse ferentiation outcome [16, 49]. Although we obtained very
RPCs where the rapid loss of rhodopsin-expressing cells was similar results from the differentiation of cells of the H9
observed shortly after plating in RPC culture [48]. How to hESC line and one hiPSC line generated in our laboratory, we
promote the long-term survival of postmitotic photoreceptors are aware that the application of our differentiation protocol
during prolonged culture remains an important challenge. It to other pluripotent stem cells may require optimization of
seems that this will not be achieved by structural means alone these signaling pathways for efficient photoreceptor genera-
[46]; however, purification of various cell types during their tion. Rapid developments in the field of induced pluripotency
peak production period may overcome this issue somewhat. are now clearing the path toward the production of patient-
derived stem cells that overcome the ethical and methodologi-
cal issues surrounding the use of embryonic derivatives [50,
10, 51, 52]. These, alongside reports of iPSC production in
CONCLUSION patients with HRD [16], combined with genetic correction
now set the stage for the future successful application of cell
Our three-step differentiation protocol was designed with the replacement therapy in patients suffering from currently incur-
aim to mimic neural and retinal development. To achieve able forms of blindness.
this, we combined serum- and feeder-free culture conditions
with known growth factors and metabolites that have been
used with some degree of success in previous hESC differen-
tiation studies [8, 12] alongside additional factors that are ACKNOWLEDGMENTS
implicated to play a role in retinal development from develop-
mental studies. To be able to understand the role of each of We are grateful to Fight for Sight UK, Prof. John Finlay,
these factors, we performed stepwise deletions of single and/ Newcastle Health Charity, Sunderland Eye Infirmary, Newcastle
or combined factors. This detailed analysis indicated that the University, UK NIHR Biomedical Research Centre for Ageing
most important components for achieving efficient photore- and Age-related disease and the Newcastle upon Tyne NHS
ceptor generation within a reasonable time frame (45 days) Hospitals Trust, Conselleria de Sanidad (Generalitat Valenciana),
are B27 and/or N2. While RPC and photoreceptor precursor and the Instituto de Salud Carlos III (Ministry of Science and
generation could be enhanced by the addition of Dkk1 and Innovation) for funding this work. We thank Dr. Owen Hughes
Noggin to B27-supplemented medium during the early stages and Ian Dimmick for their help with the flow cytometric analysis.

8 Lamba DA, Karl MO, Ware CB et al. Efficient generation of retinal


REFERENCES progenitor cells from human embryonic stem cells. Proc Natl Acad
Sci USA 2006;103:12769–12774.
9 Gong J, Sagiv O, Cai H et al. Effects of extracellular matrix and
1 Tschernutter M, Schlichtenbrede FC, Howe S et al. Long-term preser- neighboring cells on induction of human embryonic stem cells into
vation of retinal function in the RCS rat model of retinitis pigmentosa retinal or retinal pigment epithelial progenitors. Exp Eye Res 2008;86:
following lentivirus-mediated gene therapy. Gene Ther 2005;12: 957–965.
694–701. 10 Buchholz DE, Hikita ST, Rowland TJ et al. Derivation of functional
2 Rolling F, Le Meur G, Stieger K et al. Gene therapeutic prospects in retinal pigmented epithelium from induced pluripotent stem cells.
early onset of severe retinal dystrophy: Restoration of vision in Stem Cells 2009;27:2427–2434.
RPE65 Briard dogs using an AAV serotype 4 vector that specifically 11 Meyer JS, Shearer RL, Capowski EE et al. Modeling early retinal
targets the retinal pigmented epithelium. Bull Mem Acad R Med Belg development with human embryonic and induced pluripotent stem
2006;161:497–508. cells. Proc Natl Acad Sci USA 2009;106:16698–16703.
3 Bainbridge JW, Smith AJ, Barker SS et al. Effect of gene therapy on 12 Osakada F, Ikeda H, Mandai M et al. Toward the generation of rod
visual function in Leber’s congenital amaurosis. N Engl J Med 2008; and cone photoreceptors from mouse, monkey and human embryonic
358:2231–2239. stem cells. Nat Biotechnol 2008;26:215–224.
4 Maguire AM, Simonelli F, Pierce EA et al. Safety and efficacy of 13 Vugler A, Carr AJ, Lawrence J et al. Elucidating the phenomenon of
gene transfer for Leber’s congenital amaurosis. N Engl J Med 2008; HESC-derived RPE: Anatomy of cell genesis, expansion and retinal
358:2240–2248. transplantation. Exp Neurol 2008;214:347–361.
5 Ashtari M, Cyckowski LL, Monroe JF et al. The human visual cortex 14 Hirami Y, Osakada F, Takahashi K et al. Generation of retinal cells
responds to gene therapy-mediated recovery of retinal function. J Clin from mouse and human induced pluripotent stem cells. Neurosci Lett
Invest 2011;121:2160–2168. 2009;458:126–131.
6 MacLaren RE, Pearson RA, MacNeil A et al. Retinal repair by trans- 15 Klimanskaya I, Hipp J, Rezai KA et al. Derivation and comparative
plantation of photoreceptor precursors. Nature 2006;444:203–207. assessment of retinal pigment epithelium from human embryonic stem
7 Lakowski J, Baron M, Bainbridge J et al. Cone and rod photoreceptor cells using transcriptomics. Cloning Stem Cells 2004;6:217–245.
transplantation in models of the childhood retinopathy Leber congeni- 16 Meyer JS, Howden SE, Wallace KA et al. Optic vesicle-like structures
tal amaurosis using flow-sorted Crx-positive donor cells. Hum Mol derived from human pluripotent stem cells facilitate a customized
Genet 2010;19:4545–4559. approach to retinal disease treatment. Stem Cells 2011;29:1206–1218.

www.StemCells.com
686 Efficient Photoreceptor Generation from Human Stem Cells

17 Sauve Y, Klassen H, Whiteley SJ et al. Visual field loss in RCS rats and 35 Price PJ, Brewer GJ. Protocols for Neural Cell Culture. 3rd ed.
the effect of RPE cell transplantation. Exp Neurol 1998;152:243–250. Humana Press, Inc., Totowa, New Jersey, 2001: Chapter 19.
18 Lund RD, Adamson P, Sauve Y et al. Subretinal transplantation of 36 Gallego MJ, Porayette P, Kaltcheva MM et al. The pregnancy
genetically modified human cell lines attenuates loss of visual function hormones human chorionic gonadotropin and progesterone induce
in dystrophic rats. Proc Natl Acad Sci USA 2001;98:9942–9947. human embryonic stem cell proliferation and differentiation into neu-
19 Lund RD, Wang S, Klimanskaya I et al. Human embryonic stem cell- roectodermal rosettes. Stem Cell Res Ther 2010;1:28, p 1–13.
derived cells rescue visual function in dystrophic RCS rats. Cloning 37 Gaspard N, Bouschet T, Hourez R et al. An intrinsic mechanism of
Stem Cells 2006;8:189–199. corticogenesis from embryonic stem cells. Nature 2008;455:351–357.
20 Coffey PJ, Girman S, Wang SM et al. Long-term preservation of 38 Rho JY, Yu K, Han JS et al. Transcriptional profiling of the develop-
cortically dependent visual function in RCS rats by transplantation. mentally important signalling pathways in human embryonic stem
Nat Neurosci 2002;5:53–56. cells. Hum Reprod 2006;21:405–412.
20 Carr AJ, Vugler AA, Hikita ST et al. Protective effects of human iPS- 39 Pera EM, Wessely O, Li SY et al. Neural and head induction by insu-
derived retinal pigment epithelium cell transplantation in the retinal lin-like growth factor signals. Dev Cell 2001;1:655–665.
dystrophic rat. PLoS One 2009;4:e8152, p 1–12. 40 del Barco Barrantes I, Davidson G, Gr€ one HJ et al. Dkk-1 and noggin
21 Banin E, Obolensky A, Idelson M et al. Retinal incorporation and dif- cooperate in mammalian head induction. Genes Dev 2003;17:

Downloaded from https://academic.oup.com/stmcls/article/30/4/673/6415666 by CINVESTAV user on 26 May 2022


ferentiation of neural precursors derived from human embryonic stem 2239–2244.
cells. Stem Cells 2006;24:246–257. 41 Jin ZB, Okamoto S, Osakada F et al. Modeling retinal degeneration
22 Lamba DA, Gust J, Reh TA. Transplantation of human embryonic using patient-specific induced pluripotent stem cells. PLoS One 2011;
stem cell-derived photoreceptors restores some visual function in 6:e17084, p 1–8.
Crx-deficient mice. Cell Stem Cell 2009;4:73–79. 42 Zhou L, Wang W, Liu Y et al. Differentiation of induced pluripotent
23 Lamba DA, McUsic A, Hirata RK et al. Generation, purification and stem cells of Swine into rod photoreceptors and their integration into
transplantation of photoreceptors derived from human induced pluripo- the retina. Stem Cells 2011;29:972–980.
tent stem cells. PLoS One 2010;5:e8763, p 1–9. 43 Finlay BL. The developing and evolving retina: Using time to organ-
24 Jiang et al. 2011 (in press). ize form. Brain Res 2008;1192:5–16.
25 Davis AA, Matzuk MM, Reh TA. Activin A promotes progenitor 44 Morrow EM, Belliveau MJ, Cepko CL. Two phases of rod photore-
differentiation into photoreceptors in rodent retina. Mol Cell Neurosci ceptor differentiation during rat retinal development. J Neurosci 1998;
2000;15:11–21. 18:3738–3748.
26 Kelly MW, Turner JK, Reh TA. Regulation of proliferation and pho- 45 Zhu D, Deng X, Spee C et al. Polarized secretion of PEDF from
toreceptor differentiation in human fetal retinal cell cultures. Invest human embryonic stem cell-derived RPE promotes retinal progenitor
Ophthalmol Vis Sci 1995;36:1280–1289. cell survival. Invest Ophthalmol Vis Sci 2010;52:1573–1585.
27 Watanabe K, Kamiya D, Nishiyama A et al. Directed differentiation 46 Eiraku M, Takata N, Ishibashi H et al. Self-organizing optic-cup mor-
of telencephalic precursors from embryonic stem cells. Nat Neurosci phogenesis in three-dimensional culture. Nature 2011;472:51–56.
2005;8:288–296. 47 Mu~ noz-Sanjuan I, Brivanlou AH. Neural induction, the default model
28 Ikeda H, Osakada F, Watanabe K et al. Generation of Rxþ/Pax6þ and embryonic stem cells. Nat Rev Neurosci 2002;3:271–280.
neural retinal precursors from embryonic stem cells. Proc Natl Acad 48 Mansergh FC, Vawda R, Millington-Ward S et al. Loss of photorecep-
Sci USA 2005;102:11331–11336. tor potential from retinal progenitor cell cultures, despite improve-
29 Levine EM, Roelink H, Turner J et al. Sonic hedgehog promotes rod ments in survival. Exp Eye Res 2010;91:500–512.
photoreceptor differentiation in mammalian retinal cells in vitro. 49 Kattman SJ, Witty AD, Gagliardi M et al. Stage-specific optimization
J Neurosci 1997;17:6277–6288. of activin/nodal and BMP signaling promotes cardiac differentiation
30 Elliott AM, Elliott KA, Kammesheidt A. High resolution array-CGH of mouse and human pluripotent stem cell lines. Cell Stem Cell 2011;
characterization of human stem cells using a stem cell focused micro- 8:228–240.
array. Mol Biotechnol 2010;46:234–242. 50 Balasubramanian S, Babai N, Chaudhuri A et al. Non cell-autonomous
31 Ma J, Zhang J, Othersen KL et al. Expression, purification, and MALDI reprogramming of adult ocular progenitors: Generation of pluripotent
analysis of RPE65. Invest Ophthalmol Vis Sci 2001;42:1429–1435. stem cells without exogenous transcription factors. Stem Cells 2009;
32 Redmond TM, Yu S, Lee E et al. Rpe65 is necessary for production of 27:3053–3062.
11-cis-vitamin A in the retinal visual cycle. Nat Genet 1998;20:344–351. 51 Jin ZB, Okamoto S, Mandai M et al. Induced pluripotent stem cells
33 Garcia-Gonzalo FR, Belmonte JCI. Albumin-associated lipids regulate for retinal degenerative diseases: A new perspective on the challenges.
human embryonic stem cell renewal. PLoS One 2008;3:e1384, p 1–10. J Genet 2009;88:417–424.
34 Schuldiner M, Yanuka O, Itskovitz-Eldor J et al. Effects of eight 52 Altshuler D, Lo Turco JJ, Rush J et al. Taurine promotes the differen-
growth factors on the differentiation of cells derived from human em- tiation of a vertebrate retinal cell type in vitro. Development 1993;
bryonic stem cells. Proc Natl Acad Sci USA 2000;97:11307–11312. 119:1317–1328.

See www.StemCells.com for supporting information available online.

You might also like