ArticlePDF Available

Reprogramming fibroblasts into induced pluripotent stem cells with Bmi1

Authors:

Abstract and Figures

Somatic cells can be reprogrammed into induced pluripotent stem (iPS) cells by the transcription factors Oct4, Sox2, and Klf4 in combination with c-Myc. Recently, Sox2 plus Oct4 was shown to reprogram fibroblasts and Oct4 alone was able to reprogram mouse and human neural stem cells (NSCs) into iPS cells. Here, we report that Bmi1 leads to the transdifferentiation of mouse fibroblasts into NSC-like cells, and, in combination with Oct4, can replace Sox2, Klf4 and c-Myc during the reprogramming of fibroblasts into iPS cells. Furthermore, activation of sonic hedgehog signaling (by Shh, purmorphamine, or oxysterol) compensates for the effects of Bmi1, and, in combination with Oct4, reprograms mouse embryonic and adult fibroblasts into iPS cells. One- and two-factor iPS cells are similar to mouse embryonic stem cells in their global gene expression profile, epigenetic status, and in vitro and in vivo differentiation into all three germ layers, as well as teratoma formation and germline transmission in vivo. These data support that converting fibroblasts with Bmi1 or activation of the sonic hedgehog pathway to an intermediate cell type that expresses Sox2, Klf4, and N-Myc allows iPS generation via the addition of Oct4.
Characterization of iPS cells generated from MEFs by retroviral transduction with Oct4 and Bmi1. (A) Hypothesis of Bmi1’s function in the course of reprogramming MEFs into dBO- (left) and BO-iPS cell (right). (B) Phase contrast images of vehicle- (upper) and Bmi1-transduced (lower) MEFs cultured in proliferation medium (left) and in NSC medium (right). Bmi1-transduced MEFs cultured in NSC medium for 3-7 days rapidly changed morphology, resulting in bipolar and expanded NSC-like cells (right). Scale bars, 200 μ m. (C) Characterization of NSC-like cells was conducted by AP staining and immuno - cytochemistry for Nestin and Sox2, as well as RT-PCR analysis of NSC marker genes (right). Scale bars, 200 μ m. (D) NSC- like cells exhibit multipotency and thus can give rise to cells expressing neuronal and glial markers for oligodendrocytes (O4, CNPase, and GalC), neurons (Tuj1 and Map2a), and astrocytes (GFAP and S100). Scale bars, 200 μ m. (E) Phase contrast images showing mES cells (upper left) and BO-iPS cells (clone 1; lower left) on feeder cells. The reprogramming efficiency of dBO- and BO-iPS cells was quantified by determining the percentage of Nanog-positive colonies (right). (F) Characterization of BO-iPS-1 cells. AP staining, as well as SSEA1, Oct4, Sox2, and Nanog immunoreactivities, were measured in mES cells (upper panels) and BO-iPS cells (lower panels). Scale bars, 200 μ m. (G) Scatter plots of the global gene expression compar- ing BO-iPS cells with MEFs (right) and BO-iPS cells with mES cells (left). Red and green lines indicate 2-fold changes in gene expression levels. The pluripotency genes Oct4, Sox2, Nanog, c-Myc, and Klf4 are shown in red. (H) Bisulfite genomic se - quencing of Oct4 and Nanog promoter regions in mES cells, MEFs, and BO-iPS clones (1, 2, and 3). Open and filled circles indicate unmethylated and methylated CpG dinucleotides, respectively. Cell Research | Vol 21 No 9 | September 2011
… 
Generation and characterization of 1F ShO-iPS cells. (A) Hypothesis of induction of Bmi1 in the course of reprogramming. (B) Induction of sonic hedgehog target genes by Shh treatment. RT-PCR and qPCR of mRNAs from MEFs treated with vehicle (con) or Shh were analyzed for the induction of Shh target genes (Gli1, Bmi1, Sox2, and N-Myc, as well as p16 Ink4a , p19 Arf , and Klf4). Data are from a representative experiment (left) and are shown as the means and SD’s of rela - tive values compared to control MEFs ( n = 3). * P < 0.05 compared to MEFs. (C) Phase contrast images of vehicle- (shh(−)) and Shh-treated (shh(+)) MEFs cultured in proliferation medium (left) or NSC medium (right). Shh-treated MEFs cultured in NSC medium for 7 days rapidly changed morphology, resulting in bipolar (right) and expanded neurosphere-like cells (right). Scale bars, 200 μ m. (D) Characterization of neurosphere-like morphology from Shh-treated MEFs was conducted by AP staining, immunocytochemistry, and RT-PCR for Nestin, Sox2, SSEA1, Musashi1, and CD133. Scale bars, 200 μ m. (E) Timeline for ShO-iPS cell induction using Shh treatment with retroviral transduction of Oct4 (upper and left, panel). Phase contrast images showing the ESC-like morphology of ShO-iPS cells on feeder cells and the characterization of ShO-iPS cells. AP staining, as well as SSEA1, Oct4, Sox2, and Nanog immunoreactivity, was detected in ShO-iPS cells (lower panel). The reprogramming efficiency of BO- and ShO-iPS cells in reprogramming MEFs. Reprogramming efficiency was quantified by determining the percentage of Nanog-positive colonies. Scale bars, 200 μ m (upper and right panel). (F) The in vivo devel- opmental potential of ShO-iPS cells. Teratomas of ShO-iPS cells differentiated into epithelium (endoderm; left), muscle and fat (mesoderm; middle), and neural rosettes (ectoderm; right). Hematoxylin and eosin-stained sections of teratomas derived from ShO-iPS cells in a nude mouse host after 8 weeks are shown. Scale bars, 200 μ m.
… 
Content may be subject to copyright.
ORIGINAL ARTICLE
Reprogramming broblasts into induced pluripotent stem
cells with Bmi1
Jai-Hee Moon1, June Seok Heo1, Jun Sung Kim1, Eun Kyoung Jun1, 2, Jung Han Lee1, 2, Aeree Kim3,
Jonggun Kim4, Kwang Youn Whang4, Yong-Kook Kang5, Seungeun Yeo5, Hee-Joung Lim4, Dong Wook Han6,
Dong-Wook Kim7, Sejong Oh8, Byung Sun Yoon1, Hans R Schöler9, 10, Seungkwon You1
1Laboratory of Cell Function Regulation, College of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Republic
of Korea; 2Division of Stem Cell Research Institute, Stemmedience Corp., Seoul, Republic of Korea; 3Department of Pathology,
College of Medicine, Korea University Guro Hospital, Seoul, Republic of Korea; 4Division of Biotechnology, College of Life Sci-
ences and Biotechnology, Korea University, Seoul, Republic of Korea; 5Development and Differentiation Research Center, KRIBB,
Daejeon 305-333, Republic of Korea; 6Department of Stem Cell Biology, SMART Institute of Advanced Biomedical Science,
Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; 7Department of Physiology, Center for Cell
Therapy, Yonsei University College of Medicine, Seoul, Republic of Korea; 8Division of Animal Science, Chonnam National Uni-
versity, Gwangju 500-757, Republic of Korea; 9Department of Cell and Developmental Biology, Max Planck Institute for Molecu-
lar Biomedicine,Röntgenstraße 20, Münster D-48149, Germany; 10Medical Faculty, University of Münster, Domagkstr. 3, Münster
D-48149, Germany
Correspondence: Seungkwon Youa, Hans R Schölerb, Byung Sun Yoonc
aTel: +82-2-3290-3057; Fax: +82-2-3290-3507
E-mail: bioseung@korea.ac.kr
bTel: +49-251-70365-300; Fax: +49-251-70365-399
E-mail: ofce@mpi-muenster.mpg.de
cTel: +82-2-3290-3493; Fax: +82-2-3290-3507
E-mail: biosun302@korea.ac.kr
Received 9 February 2011; revised 28 March 2011; accepted 4 May 2011;
published online 28 June 2011
Somatic cells can be reprogrammed into induced pluripotent stem (iPS) cells by the transcription factors Oct4,
Sox2, and Klf4 in combination with c-Myc. Recently, Sox2 plus Oct4 was shown to reprogram broblasts and Oct4
alone was able to reprogram mouse and human neural stem cells (NSCs) into iPS cells. Here, we report that Bmi1
leads to the transdifferentiation of mouse broblasts into NSC-like cells, and, in combination with Oct4, can replace
Sox2, Klf4 and c-Myc during the reprogramming of broblasts into iPS cells. Furthermore, activation of sonic hedge-
hog signaling (by Shh, purmorphamine, or oxysterol) compensates for the effects of Bmi1, and, in combination with
Oct4, reprograms mouse embryonic and adult broblasts into iPS cells. One- and two-factor iPS cells are similar to
mouse embryonic stem cells in their global gene expression prole, epigenetic status, and in vitro and in vivo differ-
entiation into all three germ layers, as well as teratoma formation and germline transmission in vivo. These data sup-
port that converting broblasts with Bmi1 or activation of the sonic hedgehog pathway to an intermediate cell type
that expresses Sox2, Klf4, and N-Myc allows iPS generation via the addition of Oct4.
Keywords: reprogramming; transdifferentiation; neural stem cells; induced pluripotent stem cells; Bmi1; Oct4
Cell Research (2011) 21:1305-1315. doi:10.1038/cr.2011.107; published online 28 June 2011
Introduction
The potential of induced pluripotent stem (iPS) cell
technology is enormous, but comprehension of the mo-
lecular mechanisms that underlie reprogramming is mea-
ger, largely because the procedure is still very inefcient.
Recently, a number of groups have demonstrated that the
inactivation of p53 markedly increases the efciency of
iPS cell generation [1-6]. Furthermore, by reducing the
expression of both p16Ink4a and p19Arf (both of which are
encoded by alternative reading frames of the Ink4a/Arf
locus, also known as the Cdkn2a locus), iPS cell forma-
tion was increased relative to that achieved by reducing
the expression of p19Arf alone.
Bmi1 was rst identied as a proto-oncogene that co-
operates with c-Myc to promote the formation of B- and
T-cell lymphomas [7, 8] by inhibiting c-Myc-induced
npg
Cell Research (2011) 21:1305-1315.
© 2011 IBCB, SIBS, CAS All rights reserved 1001-0602/11 $ 32.00
www.nature.com/cr
Reprogramming of broblast into iPSCs with Bmi1
1306
npg
Cell Research | Vol 21 No 9 | September 2011
apoptosis through repression of the Ink4a/Arf locus [9,
10]. Bmi1 is also required for the self-renewal of stem
cells and the Ink4a/Arf locus is the main target of Bmi1
stem cell proliferation activity [11-13]. Furthermore, ac-
tivation of the sonic hedgehog (Shh) signaling pathway
induces Bmi1, Sox2, and N-Myc expression, resulting
in increased proliferation of neural precursors [14-16].
In this study, we hypothesized that cells with reduced
expression of p16Ink4a and p19Arf as well as increased N-
Myc, Klf4, and Sox2 expression [13, 16] mediated by
Bmi1 overexpression could be efciently converted into
iPS cells with either two factors (Oct4 and Sox2: hereaf-
ter designated as 2F-Bmi1-iPS cells (OSB)) or only one
factor (Oct4: hereafter designated as BO-iPS cells). Fur-
thermore, we also asked whether Shh or activation of the
Shh signaling pathway by oxysterol or purmorphamine
can regulate the downstream target genes of Bmi1 in
the generation of BO-iPS cells. Our study demonstrates
that Bmi1 has dual effects on iPS cell generation from
broblasts by both suppressing p16 Ink4a and p19 Arf and
augmenting Sox2 and N-Myc. By inducing Bmi1 with
some chemicals, we have also shown that only one
transcription factor (Oct4) is required to reprogram fi-
broblasts into pluripotent cells, which have the ability to
differentiate into all three germ layer cell types and are
capable of germline transmission.
Results
Bmi1 replaces the function of Klf4 and c-Myc, and
increases reprogramming efficiency
Considering that Bmi1 is essential for the self-renewal
of stem cells through repression of the p53 and Rb path-
ways, we investigated whether Bmi1 could enhance the
reprogramming of broblasts into iPS cells. To answer
this question, we rst determined the expression levels
of reprogramming-related genes (p16Ink4a, p19Arf, Sox2,
N-Myc, p53, and Klf4) in parental mouse embryonic
broblasts (MEFs) and Bmi1-transduced cells. p16Ink4a,
p19Arf, and p53 were significantly repressed in Bmi1-
transduced cells compared to MEFs; however, consistent
with a previous report [16], Sox2, N-Myc, and Klf4
were abundantly expressed (Figure 1A and 1B). Next,
we tested whether Bmi1 could replace Oct4, Sox2, Klf4,
or C-Myc. The four transcription factors (Oct4, Sox2,
Klf4, and C-Myc; OSKM) or a combination of three
of the transcription factors with Bmi1 (OSKB, OSBM,
OBKM, and BSKM) were introduced into MEFs. Bmi1
was able to replace Sox2, Klf4, or C-Myc in inducing
Nanog-positive colonies that resemble embryonic stem
(ES) cells (Figure 1C and Supplementary information,
Figure S1). However, we found that in the absence of
Oct4, Nanog positive colonies were not formed (data not
shown), indicating that Bmi1 is not able to replace Oct4
for reprogramming MEFs. We also tested whether Bmi1
was able to replace both Klf4 and C-Myc (Figure 1A).
We reprogrammed mouse broblasts with Oct4 and Sox2
or with the two factors plus Bmi1 (2F-Bmi1-iPS (OSB)).
Overexpression of Bmi1 in these two-factor experiments
increased the number of Nanog-positive colonies, con-
sistent with a role of Bmi1 as a replacement for Klf4 and
C-Myc, as well as a limited role in regulating the p53
and Rb pathways during reprogramming. The OSB colo-
nies were very similar to those comprised of mouse ES
cells and expressed pluripotency-associated transcription
factors as well as pluripotent cell surface markers (Figure
1D and Supplementary information, Figure S1). Taken
together, these data suggest that Bmi1-mediated regula-
tion of N-Myc, Klf4, p16Ink4a, and p19Arf activity mark-
edly increases reprogramming efciency.
Induction of fibroblasts into iPS cells with Bmi1 plus
Oct4
Methods designed to reduce the number of factors
necessary for reprogramming have taken advantage of
endogenously expressed reprogramming factors such as
Sox2 [17]. For example, studies show that adult mouse
neural stem cells (NSCs), which exhibit endogenous
Sox2 expression, can be reprogrammed by Oct4 alone [17,
18]. However, methods reprogramming somatic cells
that lack endogenous Sox2 expression to pluripotency
with Oct4 alone need to be explored. Based on previ-
ous results [16], we hypothesized that cells transduced
by Bmi1 could be transdifferentiated into NSC-like
cells and then converted into iPS cells by Oct4 (Figure
2A left panel). We rst determined whether Bmi1 could
transdifferentiate MEFs into NSC-like cells. Bmi1-
transduced MEFs, but not empty vector-transduced
MEFs, formed colonies exhibiting an NSC-like morphol-
ogy within 3-7 days in NSC culture (Figure 2A and 2B).
Of the 45 colonies generated, four were selected and
grown using standard mouse NSC culturing methods.
All four selected colonies expressed genes and cell sur-
face markers characteristic of mouse NSCs, including
Nestin and Sox2, as well as AP activity (Figure 2C). In
addition, Bmi1-transduced spheres gave rise to neurons,
oligodendrocytes, and astrocytes (Figure 2D). Next, we
investigated whether Bmi1-transduced NSC-like cells
could be reprogrammed into iPS cells by transduction
with Oct4 alone. Indeed, we succeeded in generating ES-
like colonies within 10-14 days. We refer to these repro-
grammed cells as transdifferentiated BO-iPS cells (dBO-
iPS cells). These cells were generated from MEFs that
were rst transdifferentiated into NSC-like cells and then
www.cell-research.com | Cell Research
Jai-Hee Moon et al.
1307
npg
further reprogrammed into iPS cells (Figure 2E). We also
determined whether Bmi1 could replace Sox2, Klf4, and
C-Myc and, in combination with Oct4, reprogram MEFs
into iPS cells (Figure 2A right panel). We introduced
Bmi1 and Oct4 into MEFs and were able to induce the
formation of ES-like cells, hereafter designated as BO-
iPS cells. The estimated reprogramming efciency was
calculated at 0.01% and 0.17% for dBO- and BO-iPS
cells, respectively. The reprogramming efficiency of
dBO-iPS cells was approximately 6% of that obtained
Figure 1 Increased generation of iPS cells by overexpression of Bmi1. (A) Hypothesis of Bmi1’s function in the course of re-
programming. (B) MEFs were infected with a retrovirus encoding Bmi1 or an empty vector. Three days after infection, protein
levels of Bmi1, p16Ink4a, p19Arf, and Sox2 were analyzed by western blot. Actin was used as a loading control. (C) Reprogram-
ming efciencies of factor combinations on iPS cell induction. Bmi1 replaces Sox2, Klf4, and C-Myc in the reprogramming of
MEFs. Reprogramming efciency was quantied by determining the percentage of Nanog-positive colonies. (D) MEFs were
infected with retroviruses encoding two factors (Oct4 and Sox2; OS) or with the two factors plus Bmi1 (OSB). Seven days af-
ter infection, Nanog-positive colonies formed from cells infected with OSB, but not OS (left and lower panel). Scale bars, 200
µm. Protein levels of Oct4, Sox2, c-Myc, Klf4, and p53 were analyzed by western blot. α-tubulin was used as a loading control
(right panel).
Reprogramming of broblast into iPSCs with Bmi1
1308
npg
Cell Research | Vol 21 No 9 | September 2011
Figure 2 Characterization of iPS cells generated from MEFs by retroviral transduction with Oct4 and Bmi1. (A) Hypothesis
of Bmi1’s function in the course of reprogramming MEFs into dBO- (left) and BO-iPS cell (right). (B) Phase contrast images
of vehicle- (upper) and Bmi1-transduced (lower) MEFs cultured in proliferation medium (left) and in NSC medium (right).
Bmi1-transduced MEFs cultured in NSC medium for 3-7 days rapidly changed morphology, resulting in bipolar and expanded
NSC-like cells (right). Scale bars, 200 µm. (C) Characterization of NSC-like cells was conducted by AP staining and immuno-
cytochemistry for Nestin and Sox2, as well as RT-PCR analysis of NSC marker genes (right). Scale bars, 200 µm. (D) NSC-
like cells exhibit multipotency and thus can give rise to cells expressing neuronal and glial markers for oligodendrocytes (O4,
CNPase, and GalC), neurons (Tuj1 and Map2a), and astrocytes (GFAP and S100). Scale bars, 200 µm. (E) Phase contrast
images showing mES cells (upper left) and BO-iPS cells (clone 1; lower left) on feeder cells. The reprogramming efciency of
dBO- and BO-iPS cells was quantied by determining the percentage of Nanog-positive colonies (right). (F) Characterization
of BO-iPS-1 cells. AP staining, as well as SSEA1, Oct4, Sox2, and Nanog immunoreactivities, were measured in mES cells
(upper panels) and BO-iPS cells (lower panels). Scale bars, 200 µm. (G) Scatter plots of the global gene expression compar-
ing BO-iPS cells with MEFs (right) and BO-iPS cells with mES cells (left). Red and green lines indicate 2-fold changes in gene
expression levels. The pluripotency genes Oct4, Sox2, Nanog, c-Myc, and Klf4 are shown in red. (H) Bisulte genomic se-
quencing of Oct4 and Nanog promoter regions in mES cells, MEFs, and BO-iPS clones (1, 2, and 3). Open and lled circles
indicate unmethylated and methylated CpG dinucleotides, respectively.
www.cell-research.com | Cell Research
Jai-Hee Moon et al.
1309
npg
by BO-iPS cells. The reason for the lower reprogram-
ming efficiency of dBO-iPS than BO-iPS cells is not
clear; however, it may be due to the poor transduction
efciency of Oct4 into the NSC-like spheres. To further
characterize the BO-iPS cells, six colonies that were
morphologically indistinguishable from mouse ES (mES)
cell colonies were selected and grown under standard
conditions (Figure 2E). BO-iPS cells expressed marker
genes at levels typical of mES cells (Figure 2F and Sup-
plementary information, Figures S2A, B, and S3A, B).
Furthermore, scatter plots of the global gene expression
proles showed that the BO-iPS cell prole was similar
to mES cells and was different from MEFs (Figure 2G).
To further compare BO-iPS cells with mES cells, the
methylation state of CpG dinucleotides in the Oct4 and
Nanog promoter regions was analyzed. Bisulte genomic
sequencing analyses showed that the Oct4 and Nanog
promoter regions were demethylated in BO-iPS cells
relative to the parental broblasts and showed a similar
pattern to that of mES cells (Figure 2H). Chromatin im-
munoprecipitation (ChIP) analyses showed that the Oct4,
Sox2, and Nanog promoters had increased acetylation
of histone H3 (AcH3) and decreased dimethylation of
lysine 9 of histone H3 (diMeK9H3) (Supplementary in-
formation, Figure S2C), consistent with the epigenetic
remodeling that occurs during reprogramming. Oct4
mRNA was transcribed from the endogenous Oct4 locus,
which can be distinguished from the virally expressed
human Oct4. Genomic integration of the Bmi1 and Oct4
transgenes was conrmed (Supplementary information,
Figure S3C), and the expression of both transgenes was
efciently silenced in all iPS cell lines examined (Sup-
plementary information, Figure S3D) [19].
We next investigated the differentiation potential of
BO-iPS cells in vitro with the embryoid body (EB) as-
say. EBs derived from BO-iPS cells expressed markers
of the three germ layers, including the endoderm marker
GATA4, the mesoderm markers smooth muscle actin
and Brachyury, and the ectoderm marker Nestin (Figure
3A). To investigate the differentiation potential of BO-
iPS cells in vivo, we analyzed teratomas that formed after
subcutaneous injection of BO-iPS cells into nude mice.
These teratomas contained derivatives of the three em-
bryonic germ layers, including neural epithelium, mus-
cle, cartilage, and various glandular structures (Figure 3B
and Supplementary information, Figure S3E). To assess
their developmental potential, we tested whether BO-
iPS cells could generate chimeric mice after injection
into blastocysts. According to coat color, BO-iPS clones
generated chimeric mice with germline transmission
(Figure 3C). Taken together, these results demonstrate
that Bmi1 can either transdifferentiate MEFs into NSC-
like cells and/or replace Sox2, Klf4, and C-Myc during
reprogramming of MEFs into iPS cells in the presence
of Oct4. In addition, our results suggest that chemically
induced activation of Bmi1 may be a useful strategy for
enhancing reprogramming efficiency without genetic
manipulation of the potential oncogenic Bmi1.
Shh induces Bmi1 and stimulates the generation of iPS
cells from fibroblasts by transduction with Oct4 alone
Shh, the most prominent member of the Hedgehog
family, plays an essential role during development. The
Shh signaling pathway involves the activation of Gli
transcription factors, which regulate the transcription
of target genes including Gli1 and Ptch1. Furthermore,
Bmi1, Sox2, and N-Myc expression was upregulated in
response to Shh treatment and parallels the expression of
Gli1, suppressor of fused (Sufu), and cyclin D2, which
is indicative of the activation of the Shh pathway and in-
duction of proliferation [14-16, 20, 21]. Moreover, over-
expression of Gli1 induces Bmi1 expression, suggesting
that Bmi1 is a downstream target in the Shh pathway [15].
Therefore, we tested whether Shh could replace Bmi1
in the generation of dBO- and BO-iPS cells. Bmi1, Sox2,
N-Myc, Klf4, and Gli1 mRNAs were upregulated (in
contrast to p16Ink4a and p19Arf mRNAs, which were sup-
pressed) in response to Shh treatment as early as 72 h
after incubation, indicating the activation of the Shh
pathway (Figure 4A and 4B). Moreover, cells developed
into colonies exhibiting an NSC-like morphology within
3-7 days of Shh treatment in standard NSC culture condi-
tions (Figure 4C). These NSC-like cells expressed genes
and cell surface markers characteristic of mouse NSCs,
including Sox2, Nestin, and SSEA1, as well as AP activ-
ity (Figure 4D). Shh-treated NSC-like cells were then
transduced with Oct4 to reprogram them into iPS cells
(1F combination of Shh and Oct4, hereafter designated
as ShO-iPS cells). ShO-iPS colonies obtained within 14
days in culture were further analyzed by the same tests
described above for BO-iPS cells to conrm reprogram-
ming to pluripotency (Figure 4E-F and Supplementary
information, Figure S4). Taken together, these results
demonstrate that Shh can induce Bmi1, and together with
Oct4, can reprogram MEFs into iPS cells that are very
similar to mES cells (Supplementary information, Figure
S4I).
Recently, it was demonstrated that specic oxysterol
and purmorphamine not only stimulate the Shh pathway
but also activate Shh target gene transcription through
the protein Smo [22, 23]. Similar to ShO-iPS cells, treat-
ment of MEFs with either oxysterol or purmorphamine
activated the Shh pathway, reprogramming MEFs into
NSC-like cells that exhibited gene expression profiles
Reprogramming of broblast into iPSCs with Bmi1
1310
npg
Cell Research | Vol 21 No 9 | September 2011
Figure 3 In vitro and in vivo differentiation of BO-iPS cells. (A) In vitro differentiation of BO-iPS cells. Micrographs show EBs
generated from BO-iPS-1 clones and their in vitro differentiation into ectodermal, mesodermal, and endodermal cell types,
as revealed by immunoreactivity to typical markers Nestin, Brachyury, SMA, and GATA4, respectively. Nuclei were counter-
stained with DAPI (blue). Scale bars, 200 µm. RT-PCR analysis shows that cDNAs from EBs exhibit expression of represen-
tative lineage markers in differentiating cells. (B) The in vivo developmental potential of BO-iPS cells. Teratomas generated
by BO-iPS cells differentiated into neural rosettes (ectoderm), muscle and fat (mesoderm), and epithelium (endoderm). He-
matoxylin and eosin-stained sections of teratomas derived from BO-iPS cells in a nude mouse after 8-10 weeks are shown.
Scale bars, 200 µm. (C) Chimeric mouse (upper panels) and germline contribution of BO-iPS cells in adult chimera gonads
(lower panels). Established iPS cells give rise to live chimeras (upper left) after an injection of BO-iPS cells (Balb/c genetic
background) into a C57BL6 blastocyst and contribute to the germline (lower left). PCR was performed on genomic DNA to
detect exogenous and endogenous transgene integration in chimeric (upper right) and germline transmission mice (lower
right) produced with BO-iPS cells.
www.cell-research.com | Cell Research
Jai-Hee Moon et al.
1311
npg
Figure 4 Generation and characterization of 1F ShO-iPS cells. (A) Hypothesis of induction of Bmi1 in the course of
reprogramming. (B) Induction of sonic hedgehog target genes by Shh treatment. RT-PCR and qPCR of mRNAs from MEFs
treated with vehicle (con) or Shh were analyzed for the induction of Shh target genes (Gli1, Bmi1, Sox2, and N-Myc, as well
as p16Ink4a, p19Arf, and Klf4). Data are from a representative experiment (left) and are shown as the means and SD’s of rela-
tive values compared to control MEFs (n = 3). *P < 0.05 compared to MEFs. (C) Phase contrast images of vehicle- (shh(−))
and Shh-treated (shh(+)) MEFs cultured in proliferation medium (left) or NSC medium (right). Shh-treated MEFs cultured
in NSC medium for 7 days rapidly changed morphology, resulting in bipolar (right) and expanded neurosphere-like cells
(right). Scale bars, 200 µm. (D) Characterization of neurosphere-like morphology from Shh-treated MEFs was conducted by
AP staining, immunocytochemistry, and RT-PCR for Nestin, Sox2, SSEA1, Musashi1, and CD133. Scale bars, 200 µm. (E)
Timeline for ShO-iPS cell induction using Shh treatment with retroviral transduction of Oct4 (upper and left, panel). Phase
contrast images showing the ESC-like morphology of ShO-iPS cells on feeder cells and the characterization of ShO-iPS
cells. AP staining, as well as SSEA1, Oct4, Sox2, and Nanog immunoreactivity, was detected in ShO-iPS cells (lower panel).
The reprogramming efciency of BO- and ShO-iPS cells in reprogramming MEFs. Reprogramming efciency was quantied
by determining the percentage of Nanog-positive colonies. Scale bars, 200 µm (upper and right panel). (F) The in vivo devel-
opmental potential of ShO-iPS cells. Teratomas of ShO-iPS cells differentiated into epithelium (endoderm; left), muscle and
fat (mesoderm; middle), and neural rosettes (ectoderm; right). Hematoxylin and eosin-stained sections of teratomas derived
from ShO-iPS cells in a nude mouse host after 8 weeks are shown. Scale bars, 200 µm.
Reprogramming of broblast into iPSCs with Bmi1
1312
npg
Cell Research | Vol 21 No 9 | September 2011
characteristic of NSCs (Supplementary information,
Figure S5A-S5C). Furthermore, the treatment of MEFs
with oxysterol and/or purmorphamine enhanced the
reprogramming of MEFs to pluripotency by the forced
expression of Oct4 (1F combinations of oxysterol and/or
purmorphamine and Oct4, hereafter designated as OxyO-
iPS, PO-iPS, or POxyO-iPS cells) (Supplementary in-
formation, Figure S5D). Again, the tests described above
were successfully conducted with PO-iPS and OxyO-iPS
cells (Supplementary information, Figures S5E-M and
S6A-H). Furthermore, PO-iPS cells were germline com-
petent, as demonstrated by the generation of albino off-
spring from crossing chimeric mice with wild-type mice
(Supplementary information, Figure S6I). These results
demonstrate that MEFs can be reprogrammed to pluripo-
tency by Oct4 alone when the Shh pathway is activated.
Given that Bmi1 is an important regulator of repro-
gramming-related genes (Figure 1A and 1B) [13], the
transdifferentiation of MEFs into NSC-like cells, and the
generation of iPS cells with Oct4, we studied whether
Figure 5 Decreased generation of iPS cells by the knockdown of Bmi1. (A, B) Inhibition of Bmi1 expression prevents neu-
rosphere formation. Scale bars, 200 µm. (C) qPCR analysis of Bmi1 target genes in vehicle and Bmi1 siRNA-transfected
purmorphamine- or oxysterol-treated MEFs. *P < 0.05 compared to control. (D) The reprogramming efciency of vehicle and
Bmi1 siRNA-transfected POxyO-iPS cells in reprogramming MEFs. Reprogramming efciency was quantied by determining
the percentage of Nanog-positive colonies. (E) Model summarizing the presented data. Bmi1 and Bmi1 inducers (Shh, purmor-
phamine, and oxysterol) enhance Oct4-induced reprogramming of broblasts by downregulating p16Ink4a and p19Arf expression
and upregulating Sox2, N-Myc, and Klf4 expression.
www.cell-research.com | Cell Research
Jai-Hee Moon et al.
1313
npg
knocking down Bmi1 expression would blunt neural
sphere formation. Transdifferentiation was performed
in the presence of oxysterol and/or purmorphamine to
induce the transdifferentiation of MEFs into NSC-like
cells. The formation of neurospheres and reprogram-
ming-related gene expression were signicantly altered
by the knock down of Bmi1 (Figure 5A-5C). Moreover,
when cells treated with both oxysterol and purmor-
phamine were transduced with Oct4 in the presence of
Bmi1 siRNA, the formation of Nanog-positive colonies
was signicantly decreased compared to control (Figure 5D).
We next tested the combination of oxysterol and Oct4
on the reprogramming of tail-tip broblasts (TTFs) from
10-week-old male mice (hereafter designated as OxyO-
iPS-TTF cells) and established two independent iPS
cell lines. These cells were very similar to ES cells with
an identical differentiation potential (Supplementary
information, Figure S7). Growth rates and hierarchial
clustering analysis of the global gene expression proles
showed that 1F (Oct4) and 2F (Oct4 and Bmi1) iPS cells
clustered close to mES cells and were distinct from the
parental MEFs and Bmi1-transduced neurospheres (Sup-
plementary information, Tables S1-S2 and Supplemen-
tary information, Figure S8). The chromosomal stability
of 1F and 2F iPS cells was confirmed by metaphase
spread (Supplementary information, Figure S9). Taken
together, these results show that Bmi1 promotes iPS cell
generation from broblasts; Bmi1 likely has dual effects
on iPS cell generation from broblasts, by both suppress-
ing p16Ink4a and p19Arf and augmenting Sox2 and N-Myc
(Figure 5E).
Discussion
In summary, our data support two main conclusions.
First, Bmi1 can replace Sox2, Klf4, and C-Myc during
reprogramming and enhances the reprogramming effi-
ciency of mouse broblasts infected with Oct4 and Sox2.
This suggests that not only p16Ink4a and p19Arf [1-4, 6],
but also Sox2 and C-Myc [17-19, 24] are rate-limiting
determinants in the reprogramming process. The second
conclusion relates to the number of factors applied to
somatic cells for iPS cell generation. We demonstrated
previously that mouse and human NSCs, which express
endogenous Sox2, can be reprogrammed with Oct4 alone
[17, 18]. Here, we expand upon these results by showing
that Oct4, together with Bmi1 or activators of the Shh
signaling pathway (Shh, oxysterol, and purmorphamine
in this study), is sufcient for the generation of iPS cells
from mouse embryonic and adult broblasts. This is es-
pecially crucial because adult broblasts do not express
Sox2. This study, together with others is an important
step forward in dening the critical determinants for the
generation of iPS cells from differentiated fibroblasts
[25-27]. Future studies will determine if we can combine
our direct reprogramming procedure with small molecule
compounds to activate endogenous expression of Oct4,
knock down Oct4-specific suppressor(s), or achieve
reprogramming with Oct4 recombinant protein alone.
Materials and Methods
Generation of 4F and 2F iPS cells
pBabe-based retroviral vectors encoding Oct4, Sox2, Klf4, or
C-Myc (from Gou Young Koh), and Bmi1 (from Goberdhan P.
Dimri) were transfected into PT67 amphotropic packaging cells
(Clontech) using Turbofect (Fermentas) according to the manu-
facturer’s protocol. The cells were subjected to drug selection
with 3 µg/ml puromycin (Bmi1, Klf4 and C-Myc) for 4 days or 1
000 ng/ml G418 (Sox2 and Oct4) for 14 days. The viruses were
collected after 24 h and ltered through a 0.45-µm lter before
transduction. Induction of iPS cells was performed as described
previously [24] with some modication. In brief, MEF cells (2 ×
105) were seeded in one well of a six-well plate and 2 ml of each
retroviral supernatant (Bmi1 and Oct4) was added to the cells in
the presence of 6 µg/ml of polybrene (Sigma). Two days after the
initial transduction, cells were subcultured on mitomycin C-treated
CF1 mouse feeder layers and maintained in mES cell medium
consisting of DMEM supplemented with 15% FBS (Hyclone),
β-mercaptoethanol, 1% penicillin-streptomycin, and 1 000 units/
ml leukemia inhibitory factor (LIF; Millipore). ES and iPS cells
were passaged every 3-5 days using 0.05% trypsin-EDTA and
seeded at 2 × 105 cells/well in a six-well plate. The induction of
dBO-iPS cells was performed in two steps. Generation of NSC-
like cells was achieved with MEFs infected with retroviruses
containing Bmi1, as described previously [16], and these NSC-like
cells were then further infected with retroviruses containing Oct4,
as above. Two days after Oct4-transduction, cells were grown in
mES-culture conditions as described above. For the quantication
of transduction efciency, we introduced GFP to MEF as above
and more than 70% of cells expressed GFP. To quantify efciency
of iPS generation, total number of Nanog-positive colonies were
counted and calculated as percentage of infected cell numbers,
which was calculated as GFP+ cells [28].
Generation of 1F iPS cells
To induce ShO-, PO-, OxyO-iPS, and OxyO-iPS-TTF cells,
cells were cultured in NSC medium [16] with either Shh (500 ng/
ml; R&D systems), purmorphamine (0.5, 1 µM; Calbiochem),
or 25-hydroxycholesterol (oxysterol; 0.1, 0.5 µM; Sigma) for 1
day. Next, retroviral supernatants (Oct4) in NSC medium con-
taining 6 µg/ml polybrene were added to the cells with Shh,
purmorphamine, or oxysterol. A third round of transduction was
performed and the cells were incubated for 2 days. Two days after
the initial transduction, cells were transferred and grown on mi-
tomycin C-treated MEF layers in mES cell medium. For siRNA
experiments, cells were transfected with lipofectamine RNAiMAX
(Invitrogen). Bmi1 siRNAs and control siRNAs were purchased
from Bioneer.
Reprogramming of broblast into iPSCs with Bmi1
1314
npg
Cell Research | Vol 21 No 9 | September 2011
Microarray analysis
Total RNA from MEF, mES, and iPS cells was labeled with
Cy3. Labeled RNA was hybridized to the Agilent mouse whole-
genome array (G4122F) according to the manufacturer ’s instruc-
tions. Arrays were scanned with the G2565BA Microarray Scanner
System (Agilent Technologies). All data normalization and gene
selection was performed using GeneSpring GX 7.3 (Agilent Tech-
nologies) [24].
Bisulte sequencing analysis
DNA from MEF, mES, and iPS cells was isolated using the
Genomic DNA Purification Kit (Promega). DNA was prepared
for bisulte sequencing with the EpiTect Bisulte Kit (QIAGEN).
Treated DNA was used to amplify the sequences of interest. The
primers used for promoter fragment amplification are listed in
Supplementary information, Table S3. The resulting fragments
were cloned using the pGEM-T Easy Vector (Promega) for se-
quencing and sequenced with T7 forward and SP6 reverse primers.
ChIP assay
The ChIP assay was performed on MEFs, ES cells, and iPS
cells using the EZ ChIP Kit (Millipore) according to the manufac-
turer’s instructions. Anti-acetyl H3 and anti-dimethyl K9 H3 an-
tibodies were used in this experiment. PCR primers used for real-
time PCR are listed in Supplementary information, Table S3.
Differentiation of iPS cells
iPS cells were examined by the in-vitro differentiation of EBs.
iPS cells were trypsinized and single cells were cultured in suspen-
sion with EB medium without LIF for 7 days, and EBs were then
replated onto 0.1% gelatin-coated plates. Spontaneous differentia-
tion was examined by immunostaining and RT-PCR for represen-
tative lineage-specic markers with the indicated antibodies and
primers at various timepoints (5-7 days).
Teratoma formation
iPS cells (1 × 106/mouse) were injected under the kidney cap-
sule or subcutaneously into the dorsal ank of nude mice. Eight
to ten weeks later, the mice developed teratomas, which were re-
moved, immediately rinsed with PBS, xed in 10% formalin, and
embedded in parafn. Tissue sections 5-6 µm thick were cut and
processed for hematoxylin-eosin staining.
Chimera formation and germline transmission
Four- to ve-week-old female mice (C57BL/6) were induced to
superovulate by intraperitoneal injection of 7.5 IU PMSG followed
48 h later by intraperitoneal injection of 7.5 IU hCG and mated
with a stud male mouse (C57BL/6). Blastocysts were collected
3.5 days after vaginal plug check and ushing in H-CZB medium.
Approximately 10 iPS cells then were expelled from the injection
pipette against the inner cell mass of the blastocyst. Injected blas-
tocysts were transferred into the uterine horn of 2.5 days post co-
itum (dpc) pseudopregnant CD1 female mice that had been mated
with vasectomized male mice. Chimerism was ascertained by the
contribution rate of albino coat color (from iPS cells) in black host
pups. High-contribution chimeras were mated with C57BL/6 mice
to test for germline transmission.
Statistical analysis
Data were analyzed by analysis of variants using the general
linear model procedures of the Statistical Analysis System (SAS,
9.13 PACKAGE). Data were expressed as the means ± SD. P < 0.05
was considered signicant.
Accession codes
Gene Expression Omnibus (GEO): GSE24208
Further details and other methods can be found in the supple-
mentary information, Data S1.
Acknowledgments
We are grateful to Drs Goberdhan P Dimri (NorthShore Univer-
sity HealthSystem Research Institute, USA)and Gou Young Koh
(Korea Advanced Institute of Science and Technology, Republic of
Korea) for kindly providing the pBabe-Bmi1, pMX-Oct4, pMX-
Sox2, pMX-Klf4, and pMX-C-Myc constructs. We thank Jihyun
Kim, Jihye Hwang, and Suhyun Kwon for technical support. This
research was supported by a grant (SC-5150) from the Stem Cell
Research Center of the 21st Century Frontier Research Program
funded by the Ministry of Education, Science and Technology,
Republic of Korea, a grant (09172KFDA653) from the Korea
Food and Drug Administration, and a grant (2010-0020347)
from National Research Foundation (NRF) funded by the Korea
government (MEST). Work in Germany was supported by the
Max Planck Society and the Federal Ministry of Education and
Research (BMBF) on Cell-Based Regenerative Medicine (Grant
01GN0539).
References
1 Hong H, Takahashi K, Ichisaka T, et al. Suppression of indu-
ced pluripotent stem cell generation by the p53-p21 pathway.
Nature 2009; 460:1132-1135.
2 Kawamur a T, Suzuki J, Wang YV, et al. Linking the p53
tumour suppressor pathway to somatic cell reprogramming.
Nature 2009; 460:1140-1144.
3 Li H, Collado M, Villasante A, et al. The Ink4/Arf locus is a
barrier for iPS cell reprogramming. Nature 2009; 460:1136-
1139.
4 Marion RM, Strati K, Li H, et al. A p53-mediated DNA dama-
ge response limits reprogramming to ensure iPS cell genomic
integrity. Nature 2009; 460:1149-1153.
5 Pei D. Regulation of pluripotency and reprogramming by
transcription factors. J Biol Chem 2009; 284:3365-3369.
6 Utikal J, Polo JM, Stadtfeld M, et al. Immortalization eli-
minates a roadblock during cellular reprogramming into iPS
cells. Nature 2009; 460:1145-1148.
7 Haupt Y, Alexander WS, Barri G, Klinken SP, Adams JM. No-
vel zinc nger gene implicated as myc collaborator by retro-
virally accelerated lymphomagenesis in E mu-myc transgenic
mice. Cell 1991; 65:753-763.
8 van Lohuizen M, Verbeek S, Scheijen B, Wientjens E, van der
Gulden H, Berns A. Identication of cooperating oncogenes
in E mu-myc transgenic mice by provirus tagging. Cell 1991;
65:737-752.
9 Jacobs JJ, Kieboom K, Marino S, DePinho RA, van Lohuizen
M. The oncogene and Polycomb-group gene bmi-1 regulates
cell proliferation and senescence through the ink4a locus. Na-
www.cell-research.com | Cell Research
Jai-Hee Moon et al.
1315
npg
ture 1999; 397:164-168.
10 Jacobs JJ, Scheijen B, Voncken JW, Kieboom K, Berns A, van
Lohuizen M. Bmi-1 collaborates with c-Myc in tumorigenesis
by inhibiting c-Myc-induced apoptosis via INK4a/ARF. Ge-
nes Dev 1999; 13:2678-2690.
11 Molofsky AV, Pardal R, Iwashita T, Park IK, Clarke MF,
Morrison SJ. Bmi-1 dependence distinguishes neural stem
cell self-renewal from progenitor proliferation. Nature 2003;
425:962-967.
12 Park IK, Qian D, Kiel M, et al. Bmi-1 is required for mainte-
nance of adult self-renewing haematopoietic stem cells. Natu-
re 2003; 423:302-305.
13 Valk-Lingbeek ME, Bruggeman SW, van Lohuizen M.
Stem cells and cancer; the polycomb connection. Cell 2004;
118:409-418.
14 Hatton BA, Knoepfler PS, Kenney AM, et al. N-myc is an
essential downstream effector of Shh signaling during both
normal and neoplastic cerebellar growth. Cancer Res 2006;
66:8655-8661.
15 Leung C, Lingbeek M, Shakhova O, et al. Bmi1 is essential
for cerebellar development and is overexpressed in human
medulloblastomas. Nature 2004; 428:337-341.
16 Moon JH, Yoon BS, Kim B, et al. Induction of neural stem
cell-like cells (NSCLCs) from mouse astrocytes by Bmi1.
Biochem Biophys Res Commun 2008; 371:267-272.
17 Kim JB, Sebastiano V, Wu G, et al. Oct4-induced pluripoten-
cy in adult neural stem cells. Cell 2009; 136:411-419.
18 Kim JB, Greber B, Arauzo-Bravo MJ, et al. Direct reprogra-
mming of human neural stem cells by OCT4. Nature 2009;
461:649-643.
19 Huangfu D, Osafune K, Maehr R, et al. Induction of pluri-
potent stem cells from primary human broblasts with only
Oct4 and Sox2. Nat Biotechnol 2008; 26:1269-1275.
20 Dahmane N, Ruiz i Altaba A. Sonic hedgehog regulates the
growth and patterning of the cerebellum. Development 1999;
126:3089-3100.
21 Wechsler-Reya RJ, Scott MP. Control of neuronal precursor
proliferation in the cerebellum by Sonic Hedgehog. Neuron
1999; 22:103-114.
22 Corcoran RB, Scott MP. Oxysterols stimulate Sonic hedge-
hog signal transduction and proliferation of medulloblastoma
cells. Proc Natl Acad Sci USA 2006; 103:8408-8413.
23 Lipinski RJ, Gipp JJ, Zhang J, Doles JD, Bushman W. Unique
and complimentary activities of the Gli transcription factors
in Hedgehog signaling. Exp Cell Res 2006; 312:1925-1938.
24 Takahashi K, Yamanaka S. Induction of pluripotent stem cells
from mouse embryonic and adult broblast cultures by de-
ned factors. Cell 2006; 126:663-676.
25 Chen J, Liu J, Yang J, et al. BMPs functionally replace Klf4
and support efcient reprogramming of mouse broblasts by
Oct4 alone. Cell Res 2011; 21:205-212.
26 Li Y, Zhang Q, Yin X, et al. Generation of iPSCs from mouse
broblasts with a single gene, Oct4, and small molecules. Cell
Res 2011; 21:196-204.
27 Yuan X, Wan H, Zhao X, Zhu S, Zhou Q, Ding S. Combined
chemical treatment enables Oct4-induced reprogramming
from mouse embryonic broblasts. Stem Cells 2011; 29:549-
553.
28 Shao L, Feng W, Sun Y et al. Generation of iPS cells using
dened factors linked via the self-cleaving 2A sequences in a
single open reading frame. Cell Res 2009; 19:296-306.
(Supplementary information is linked to the online version of
the paper on the Cell Research website.)
... Other Dot1L inhibitors (SGC0946 or EPZ5676) increased the binding of SOX2 and OCT4 (or Oct6, 7,8,9) to OCT4 and NANOG enhancer regions (Figure 1), providing an epigenetically permissive state for pluripotency reprogramming [38]. Oct4 overexpression alone was sufficient to generate iPSCs when fibroblasts were treated with several molecules such as protein arginine methyltransferase inhibitor AMI-5 and TGF-β inhibitor A-83-01 [39], BMPs [40], Bmi [41], and the combination of VPA, GSK3-β inhibitor (CHIR99021), RepSox, and the Lysine-specific demethylase 1 (LSD1) inhibitor (Tranylcypromine) [42]. These successful examples of the use of chemical cues for pluripotency reprogramming demonstrate the power of small molecules as surrogates for genetic cues. ...
Article
Full-text available
Although lineage reprogramming from one cell type to another is becoming a breakthrough technology for cell-based therapy, several limitations remain to be overcome, including the low conversion efficiency and subtype specificity. To address these, many studies have been conducted using genetics, chemistry, physics, and cell biology to control transcriptional networks, signaling cascades, and epigenetic modifications during reprogramming. Here, we summarize recent advances in cellular reprogramming and discuss future directions.
... Recently studies also indicated that iPSCs generation only uses Oct4 and Sox2 without oncogenes involvement [2,3]. Further, Bmi1 can replace Sox2, Klf4, and C-Myc during reprogramming, but Bmi1 was identified as a proto-oncogene that cooperates with c-Myc to promote the formation of B-and T-cell lymphomas [4]. It has been demonstrated that the Oct4 factor alone can reprogram human neural progenitor cells to pluripotency without the assistance of additional factors [5]. ...
Article
Full-text available
The iPS cells were discovered in 2006. With their ability to differentiate into cells of all three germ layers, iPS cells have great potential for clinical applications. Oct4, Sox2, c-Myc, and Klf4 were identified as the most effective factors for generating iPS cells. Despite this, iPS cells manufactured with these factors would still be inefficient. As a member of the chromobox family, chromobox protein homolog 7 (Cbx7) binds to PRC1 and PRC2 to inhibit genes involved in differentiation. A decrease in the expression of Cbx7 is observed during embryonic stem cell differentiation. Currently, no report discusses the role of Cbx7 in the production of iPS cells. In this study, we hypothesized that Cbx7 could increase iPS cell generation. We confirmed that Cbx7 is highly expressed in pluripotent stem cells (including ES and iPS cells). In addition, transfecting Cbx7 into fibroblasts increased Oct4, Sox2, c-Myc, and Klf4 expression. Moreover, we describe a novel approach to producing iPS cells using Cbx7 in combination with Oct4, Sox2, c-Myc, and Klf4. In summary, we have demonstrated that Cbx7 enhances the reprogramming of iPS cells and characterized the stemness and pluripotency of iPS cells.
... Methods not involving transcription factors have also been developed and include the RNA-binding protein LIN28A which, when coexpressed with NANOG, OCT4, and SOX2, can replace KLF4 and c-MYC [96]. The epigenetic modifier and corepressor RCOR2 can replace SOX2 [155], and the DNA demethylase TET1, or the epithelial junction molecule E-CADHERIN, can both replace OCT4 [62,160], while one of JHDM1B or BMI1 in combination with OCT4 can reprogram cells [24,129]. OCT4 fused to the VP16 domain can also reprogram somatic cells alone [29]. These lines of research have culminated in two completely non-OSKM reprogramming cocktails, the first consisting of combinations of ESRRB, SALL4, LIN28A, DPPA2, and NANOG [36] and the second consisting of JDP2, JHDM1B, ID1/ID3, GLIS1, SALL4, and LRH1 [95].[ 1 0 _ T D $ D I F F ] In addition, suppressing DOT1L or knocking down p53 can replace c-MYC and KLF4 [93,161], and the knockdown of Dlx3 can replace OCT4 [42]. ...
... Research advances in direct cellular reprogramming offer the possibility of developing patient-speci c cellular therapies. Induced neural stem cells (iNSCs) have been derived by reprogramming somatic cells with multiple transcription factors and other methods to avoid the pluripotent phase of induced pluripotent stem cells and the risks associated with low differentiation e ciency and carcinogenicity [18][19][20][21][22][23][24][25][26][27]. Jing Nai-Ho et al used peripheral blood mononuclear cells (PBMC) reprogrammed as induced NSCs to rescue AD cognitive de cits by strengthening synaptic networks. ...
Preprint
Full-text available
Background: Neural stem cells (NSC) are essential for maintaining tissue homeostasis and promoting longevity in living organisms. As a promising approach to treating neurodegenerative diseases, NSC transplantation has been hampered by crucial issues such as cellular senescence, immune rejection, and low cell viability. Methods: MiR-302a was used to reprogram human and mouse fibroblast cells into induced neural stem cells (iNSCs). In vitro, differentiation experiments were performed to demonstrate that iNSCs have the ability to differentiate into neurons, astrocytes, and oligodendrocytes. INSCs were transplanted into nude mice to evaluate cell survival, differentiation, and tumor formation in vivo. Multi-electrode arrays were used to determine that the differentiated neurons from iNSCs have mature electrophysiological functions. INSCs were treated with oxidative damage to test their antioxidant and anti-aging abilities. The supernatant of iNSCs was used to treat aged cells to determine their antioxidant and anti-aging effects. INSCs were transplanted into SAMP8 rapid aging Alzheimer's disease (AD) mouse model for behavioral tests to evaluate the improvement and therapeutic effects of iNSCs treatment on cognitive function and memory. Tests were also performed to assess lifespan extension, improved glycemic control, promoted motor ability recovery, improved reproductive ability, and improved hearing. Results: We report that a single miR-302 factor alone can effectively reprogram human and mouse fibroblasts directly into iNSCs within 2-3 days, confirmed by cell phenotype, molecular characterization, and functional analysis. The anti-aging factors Nrf2, Sirt6, and Foxo3 are highly expressed in induced neural stem cells reprogrammed by miR-302a (miR-302a-hiNSCs). Compared to other iNSCs, miR-302a-hiNSCs showed delayed aging and increased resilience to oxidative stress. MiR-302a-hiNSCs were implanted into SAMP8 mice to improve cognition, extend longevity by 40.625%, increase fatigue resistance, and enhance blood sugar control, hair regrowth, and reproduction. Conclusion: Our study highlights the potential of iNSCs generated based on miR-302a as a promising therapeutic approach for treating various age-related diseases and conditions. We found the iNSCs treatment to improve lifespan, cognitive abilities in late-stage AD, fatigue resistance, hair regeneration, blood glucose, and fat metabolism, renal function, reproductive function, and hearing loss.
... Moreover, 1′-O-methyl-averantin targets the SHH signaling pathway via Gli1, independently of SMO. SHH signaling increases Bmi-1 expression, which in turn drives cellular reprogramming 57,58 . In particular, the downstream transcription factor Gli within the SHH pathway induces expression of Bmi-1 59-61 . ...
Article
Full-text available
Endolichenic fungi are host organisms that live on lichens and produce a wide variety of secondary metabolites. Colorectal cancer stem cells are capable of self-renewal and differentiation into cancer cells, which makes cancers difficult to eradicate. New alternative therapeutics are needed to inhibit the growth of tumor stem cells. This study examined the ability of an extract of Jackrogersella sp. EL001672 (derived from the lichen Cetraria sp.) and the isolated compound 1′-O-methyl-averantin to inhibit development of cancer stemness. The endolichenic fungus Jackrogersella sp. EL001672 (KACC 83021BP), derived from Cetraria sp., was grown in culture medium. The culture broth was extracted with acetone to obtain a crude extract. Column chromatography and reverse-phase HPLC were used to isolate an active compound. The anticancer activity of the extract and the isolated compound was evaluated by qRT-PCR and western blotting, and in cell viability, spheroid formation, and reporter assays. The acetone extract of EL001672 did not affect cell viability. However, 1′-O-methyl-averantin showed cytotoxic effects against cancer cell lines at 50 μg/mL and 25 μg/mL. Both the crude extract and 1′-O-methyl-averantin suppressed spheroid formation in CRC cell lines, and downregulated expression of stemness markers ALDH1, CD44, CD133, Lgr-5, Msi-1, and EphB1. To further characterize the mechanism underlying anti-stemness activity, we examined sonic Hedgehog and Notch signaling. The results showed that the crude extract and the 1′-O-methyl-averantin inhibited Gli1, Gli2, SMO, Bmi-1, Notch-1, Hes-1, and the CSL complex. Consequently, an acetone extract and 1′-O-methyl-averantin isolated from EL001672 suppresses colorectal cancer stemness by regulating the sonic Hedgehog and Notch signaling pathways.
... Research advances in direct cellular reprogramming offer the possibility of developing patient-specific cellular therapies. Induced neural stem cells (iNSCs) have been derived by reprogramming somatic cells with multiple transcription factors and other methods to avoid the pluripotent phase of induced pluripotent stem cells and the risks associated with low differentiation efficiency and carcinogenicity (18)(19)(20)(21)(22)(23)(24)(25)(26). Jing Nai-Ho et al used PB MNC cells reprogrammed as induced neural stem cells to rescue AD cognitive deficits by strengthening synaptic networks. ...
Preprint
Neural stem cells play a vital role in maintaining tissue stability and extending lifespan. Transplanting these cells to treat neurodegenerative diseases faces challenges like cellular aging, low viability, and immune rejection. We have effectively reprogrammed human fibroblasts into induced neural stem cells (iNSCs) via a single-factor miR-302a strategy, which converted skin fibroblasts into human-induced neural stem cells (hiNSCs) within 2-3 days. These cells showed delayed aging and increased resistance to oxidative stress compared to wild-type cells. Implanting them into the hippocampus of senescence-accelerated mice improved cognitive performance in severe Alzheimer's, prolonged lifespan by 34%, increased fatigue resistance, and improved hair regeneration and reproductive capacity. Our findings suggest that miR-302a-hiNSCs can improve functional recovery in Alzheimer's and promote healthy aging.
... Supplementary Materials: The following supporting information can be downloaded at: https: //www.mdpi.com/article/10.3390/ijms24031917/s1. References are cited in [91][92][93]. ...
Article
Full-text available
Induced pluripotent stem cells (iPSCs) can differentiate into all types of cells and can be used in livestock for research on biological development, genetic breeding, and in vitro genetic resource conservation. The Bactrian camel is a large domestic animal that inhabits extreme environments and holds value in the treatment of various diseases and the development of the local economy. Therefore, we transferred four mouse genes (Oct4, Sox2, Klf4, and c-Myc) into Bactrian camel fetal fibroblasts (BCFFs) using retroviruses with a large host range to obtain Bactrian camel induced pluripotent stem cells (bciPSCs). They were comprehensively identified based on cell morphology, pluripotency gene and marker expression, chromosome number, transcriptome sequencing, and differentiation potential. The results showed the pluripotency of bciPSCs. However, unlike stem cells of other species, late formation of stem cell clones was observed; moreover, the immunofluorescence of SSEA1, SSEA3, and SSEA4 were positive, and teratoma formation took four months. These findings may be related to the extremely long gestation period and species specificity of Bactrian camels. By mining RNA sequence data, 85 potential unique pluripotent genes of Bactrian camels were predicted, which could be used as candidate genes for the production of bciPSC in the future. Among them, ASF1B, DTL, CDCA5, PROM1, CYTL1, NUP210, Epha3, and SYT13 are more attractive. In conclusion, we generated bciPSCs for the first time and obtained their transcriptome information, expanding the iPSC genetic information database and exploring the applicability of iPSCs in livestock. Our results can provide an experimental basis for Bactrian camel ESC establishment, developmental research, and genetic resource conservation.
... PcG proteins can substitute for classical OSKM reprogramming factors. For example, the combination of Bmi1 and Oct4 can reprogramme fibroblasts into iPSCs (Moon et al., 2011) and the efficiency of iPSC reprogramming is enhanced by Ezh2 expression (Ding et al., 2014). There are thousands of PcG target loci in the genome, many of which overlap with CpG islands (CGIs) that are generally hypomethylated (Ku et al., 2008). ...
Article
Full-text available
‘Age reprogramming’ refers to the process by which the molecular and cellular pathways of a cell that are subject to age-related decline are rejuvenated without passage through an embryonic stage. This process differs from the rejuvenation observed in differentiated derivatives of induced pluripotent stem cells, which involves passage through an embryonic stage and loss of cellular identity. Accordingly, the study of age reprogramming can provide an understanding of how ageing can be reversed while retaining cellular identity and the specialised function(s) of a cell, which will be of benefit to regenerative medicine. Here, we highlight recent work that has provided a more nuanced understanding of age reprogramming and point to some open questions in the field that might be explored in the future.
... ( Supplementary Fig. 1a, b). To facilitate the reprogramming process, we employed transcription factors that have the strongest effect on direct reprogramming [19][20][21][22] . Surprisingly, we observed small round cells with a bipolar morphology consistent with OPCs in BJ and expression of GFP in SOX10::eGFP fibroblasts at 7 days post-transduction of OCT4 ( Supplementary Fig. 1c). ...
Article
Full-text available
The generation of human oligodendrocyte progenitor cells (OPCs) may be therapeutically valuable for human demyelinating diseases such as multiple sclerosis. Here, we report the direct reprogramming of human somatic cells into expandable induced OPCs (iOPCs) using a combination of OCT4 and a small molecule cocktail. This method enables generation of A2B5+ (an early marker for OPCs) iOPCs within 2 weeks retaining the ability to differentiate into MBP-positive mature oligodendrocytes. RNA-seq analysis revealed that the transcriptome of O4+ iOPCs was similar to that of O4+ OPCs and ChIP-seq analysis revealed that putative OCT4-binding regions were detected in the regulatory elements of CNS development-related genes. Notably, engrafted iOPCs remyelinated the brains of adult shiverer mice and experimental autoimmune encephalomyelitis mice with MOG-induced 14 weeks after transplantation. In conclusion, our study may contribute to the development of therapeutic approaches for neurological disorders, as well as facilitate the understanding of the molecular mechanisms underlying glial development.
... Since Bmi1 and Mll1 belong to two complexes with opposite functions for methylation, it is rather intriguing that both can enhance reprogramming. More interestingly, both Bmi1 and Mll1 are involved in not only cardiac reprogramming, but also iPSC [31] and pluripotent stem cell reprogramming [32]. These results imply that Bmi1 and Mll1 might have more general targets instead of the specific cardiogenic loci. ...
Article
Full-text available
Direct reprogramming of fibroblasts into CM-like cells has emerged as an attractive strategy to generate induced CMs (iCMs) in heart regeneration. However, low conversion rate, poor purity, and the lack of precise conversion of iCMs are still present as significant challenges. In this review, we summarize the recent development in understanding the molecular mechanisms of cardiac reprogramming with various strategies to achieve more efficient iCMs. reprogramming. Specifically, we focus on the identified critical roles of transcriptional regulation, epigenetic modification, signaling pathways from the cellular microenvironment, and cell cycling regulation in cardiac reprogramming. We also discuss the progress in delivery system optimization and cardiac reprogramming in human cells related to preclinical applications. We anticipate that this will translate cardiac reprogramming-based heart therapy into clinical applications. In addition to optimizing the cardiogenesis related transcriptional regulation and signaling pathways, an important strategy is to modulate the pathological microenvironment associated with heart injury, including inflammation, pro-fibrotic signaling pathways, and the mechanical properties of the damaged myocardium. We are optimistic that cardiac reprogramming will provide a powerful therapy in heart regenerative medicine.
Article
Full-text available
Generation of induced pluripotent stem cells by defined factors has become a useful model to investigate the mechanism of reprogramming and cell fate determination. However, the precise mechanism of factor-based reprogramming remains unclear. Here, we show that Klf4 mainly acts at the initial phase of reprogramming to initiate mesenchymal-to-epithelial transition and can be functionally replaced by bone morphogenetic proteins (BMPs). BMPs boosted the efficiency of Oct4/Sox2-mediated reprogramming of mouse embryonic fibroblasts (MEFs) to ∼1%. BMPs also promoted single-factor Oct4-based reprogramming of MEFs and tail tibial fibroblasts. Our studies clarify the contribution of Klf4 in reprogramming and establish Oct4 as a singular setter of pluripotency in differentiated cells.
Article
Full-text available
The introduction of four transcription factors Oct4, Klf4, Sox2 and c-Myc by viral transduction can induce reprogramming of somatic cells into induced pluripotent stem cells (iPSCs), but the use of iPSCs is hindered by the use of viral delivery systems. Chemical-induced reprogramming offers a novel approach to generating iPSCs without any viral vector-based genetic modification. Previous reports showed that several small molecules could replace some of the reprogramming factors although at least two transcription factors, Oct4 and Klf4, are still required to generate iPSCs from mouse embryonic fibroblasts. Here, we identify a specific chemical combination, which is sufficient to permit reprogramming from mouse embryonic and adult fibroblasts in the presence of a single transcription factor, Oct4, within 20 days, replacing Sox2, Klf4 and c-Myc. The iPSCs generated using this treatment resembled mouse embryonic stem cells in terms of global gene expression profile, epigenetic status and pluripotency both in vitro and in vivo. We also found that 8 days of Oct4 induction was sufficient to enable Oct4-induced reprogramming in the presence of the small molecules, which suggests that reprogramming was initiated within the first 8 days and was independent of continuous exogenous Oct4 expression. These discoveries will aid in the future generation of iPSCs without genetic modification, as well as elucidating the molecular mechanisms that underlie the reprogramming process.
Article
Full-text available
Induced pluripotent stem (iPS) cells have been generated from mouse and human somatic cells by ectopic expression of four transcription factors (OCT4 (also called POU5F1), SOX2, c-Myc and KLF4). We previously reported that Oct4 alone is sufficient to reprogram directly adult mouse neural stem cells to iPS cells. Here we report the generation of one-factor human iPS cells from human fetal neural stem cells (one-factor (1F) human NiPS cells) by ectopic expression of OCT4 alone. One-factor human NiPS cells resemble human embryonic stem cells in global gene expression profiles, epigenetic status, as well as pluripotency in vitro and in vivo. These findings demonstrate that the transcription factor OCT4 is sufficient to reprogram human neural stem cells to pluripotency. One-factor iPS cell generation will advance the field further towards understanding reprogramming and generating patient-specific pluripotent stem cells.
Article
Full-text available
Induced pluripotent stem (iPS) cells can be generated from somatic cells by the introduction of Oct3/4 (also known as Pou5f1), Sox2, Klf4 and c-Myc, in mouse and in human. The efficiency of this process, however, is low. Pluripotency can be induced without c-Myc, but with even lower efficiency. A p53 (also known as TP53 in humans and Trp53 in mice) short-interfering RNA (siRNA) was recently shown to promote human iPS cell generation, but the specificity and mechanisms remain to be determined. Here we report that up to 10% of transduced mouse embryonic fibroblasts lacking p53 became iPS cells, even without the Myc retrovirus. The p53 deletion also promoted the induction of integration-free mouse iPS cells with plasmid transfection. Furthermore, in the p53-null background, iPS cells were generated from terminally differentiated T lymphocytes. The suppression of p53 also increased the efficiency of human iPS cell generation. DNA microarray analyses identified 34 p53-regulated genes that are common in mouse and human fibroblasts. Functional analyses of these genes demonstrate that the p53-p21 pathway serves as a barrier not only in tumorigenicity, but also in iPS cell generation.
Article
Full-text available
The overexpression of defined transcription factors in somatic cells results in their reprogramming into induced pluripotent stem (iPS) cells. The extremely low efficiency and slow kinetics of in vitro reprogramming suggest that further rare events are required to generate iPS cells. The nature and identity of these events, however, remain elusive. We noticed that the reprogramming potential of primary murine fibroblasts into iPS cells decreases after serial passaging and the concomitant onset of senescence. Consistent with the notion that loss of replicative potential provides a barrier for reprogramming, here we show that cells with low endogenous p19(Arf) (encoded by the Ink4a/Arf locus, also known as Cdkn2a locus) protein levels and immortal fibroblasts deficient in components of the Arf-Trp53 pathway yield iPS cell colonies with up to threefold faster kinetics and at a significantly higher efficiency than wild-type cells, endowing almost every somatic cell with the potential to form iPS cells. Notably, the acute genetic ablation of Trp53 (also known as p53) in cellular subpopulations that normally fail to reprogram rescues their ability to produce iPS cells. Our results show that the acquisition of immortality is a crucial and rate-limiting step towards the establishment of a pluripotent state in somatic cells and underscore the similarities between induced pluripotency and tumorigenesis.
Article
Full-text available
The reprogramming of differentiated cells to pluripotent cells (induced pluripotent stem (iPS) cells) is known to be an inefficient process. We recently reported that cells with short telomeres cannot be reprogrammed to iPS cells despite their normal proliferation rates, probably reflecting the existence of 'reprogramming barriers' that abort the reprogramming of cells with uncapped telomeres. Here we show that p53 (also known as Trp53 in mice and TP53 in humans) is critically involved in preventing the reprogramming of cells carrying various types of DNA damage, including short telomeres, DNA repair deficiencies, or exogenously inflicted DNA damage. Reprogramming in the presence of pre-existing, but tolerated, DNA damage is aborted by the activation of a DNA damage response and p53-dependent apoptosis. Abrogation of p53 allows efficient reprogramming in the face of DNA damage and the generation of iPS cells carrying persistent DNA damage and chromosomal aberrations. These observations indicate that during reprogramming cells increase their intolerance to different types of DNA damage and that p53 is critical in preventing the generation of human and mouse pluripotent cells from suboptimal parental cells.
Article
Recently, Bmi1 was shown to control the proliferation and self-renewal of neural stem cells (NSCs). In this study, we demonstrated the induction of NSC-like cells (NSCLCs) from mouse astrocytes by Bmi1 under NSC culture conditions. These NSCLCs exhibited the morphology and growth properties of NSCs, and expressed NSC marker genes, including nestin, CD133, and Sox2. In vitro differentiation of NSCLCs resulted in differentiated cell populations containing astrocytes, neurons, and oligodendrocytes. Following treatment with histone deacetylase inhibitors (trichostatin A and valproic acid), the potential of NSCLCs for proliferation, dedifferentiation, and self-renewal was significantly inhibited. Our data indicate that multipotent NSCLCs can be generated directly from astrocytes by the addition of Bmi1.
Article
Nature is the international weekly journal of science: a magazine style journal that publishes full-length research papers in all disciplines of science, as well as News and Views, reviews, news, features, commentaries, web focuses and more, covering all branches of science and how science impacts upon all aspects of society and life.
Article
It has been established that exogenous expression of four transcription factors (Oct4, Klf4, Sox2, and c-Myc) can reprogram mammalian somatic cells to pluripotent states. Further studies demonstrated that such induced pluripotent stem cells (iPSCs) could be generated with fewer exogenous transcription factors, facilitated by endogenous expression of reprogramming factors and/or synthetic small molecules. Here, we reported identification of a new small molecule, a protein arginine methyltransferase inhibitor AMI-5, which enabled Oct4-induced reprogramming of mouse embryonic fibroblasts in combination with transforming growth factor (TGF)-β inhibitor A-83-01. The Oct4-induced iPSCs were shown similar to mouse embryonic stem cells with respect to typical pluripotency criteria. More importantly, they were shown to give rise to liveborn pups through tetraploid complementation assays, demonstrating the high quality of full reprogramming induced by this condition. Furthermore, this study suggests that regulation of protein arginine methylation might be involved in the reprogramming process.
Article
The mechanisms involved in the reprogramming of differentiated cells into induced pluripotent stem (iPS) cells by the three transcription factors Oct4 (also known as Pou5f1), Klf4 and Sox2 remain poorly understood. The Ink4/Arf locus comprises the Cdkn2a-Cdkn2b genes encoding three potent tumour suppressors, namely p16(Ink4a), p19(Arf) and p15(Ink4b), which are basally expressed in differentiated cells and upregulated by aberrant mitogenic signals. Here we show that the locus is completely silenced in iPS cells, as well as in embryonic stem (ES) cells, acquiring the epigenetic marks of a bivalent chromatin domain, and retaining the ability to be reactivated after differentiation. Cell culture conditions during reprogramming enhance the expression of the Ink4/Arf locus, further highlighting the importance of silencing the locus to allow proliferation and reprogramming. Indeed, the three factors together repress the Ink4/Arf locus soon after their expression and concomitant with the appearance of the first molecular markers of 'stemness'. This downregulation also occurs in cells carrying the oncoprotein large-T, which functionally inactivates the pathways regulated by the Ink4/Arf locus, thus indicating that the silencing of the locus is intrinsic to reprogramming and not the result of a selective process. Genetic inhibition of the Ink4/Arf locus has a profound positive effect on the efficiency of iPS cell generation, increasing both the kinetics of reprogramming and the number of emerging iPS cell colonies. In murine cells, Arf, rather than Ink4a, is the main barrier to reprogramming by activation of p53 (encoded by Trp53) and p21 (encoded by Cdkn1a); whereas, in human fibroblasts, INK4a is more important than ARF. Furthermore, organismal ageing upregulates the Ink4/Arf locus and, accordingly, reprogramming is less efficient in cells from old organisms, but this defect can be rescued by inhibiting the locus with a short hairpin RNA. All together, we conclude that the silencing of Ink4/Arf locus is rate-limiting for reprogramming, and its transient inhibition may significantly improve the generation of iPS cells.