ArticlePDF AvailableLiterature Review

Embryonic Stem Cells in Cattle

Authors:
  • INRAE (Institut National de Recherche pour l'Agriculture l'Alimentation et l'Environnement)

Abstract and Figures

Contents Because of the potential use of embryonic stem cells (ESC), especially for genetic modifications, there is great interest in establishing domestic animals‐related ESCs. Unfortunately, despite considerable efforts, validated ESC lines in species other than mice and primates are yet to be isolated. In this paper, we will summarize the current knowledge on bovine ESCs in an attempt to understand why derivation of domestic animal ESC is still unsuccessful and we will discuss some promising future approaches.
Content may be subject to copyright.
Review Article
Embryonic Stem Cells in Cattle
M Mun
˜oz
1
,CDı
´ez
1
, JN Caaman
˜o
1
, A Jouneau
2,3,4
, I Hue
2,3,4
and E Go
´mez
1
1
Area de Gene
´tica y Reproduccio
´n, SERIDA, Camino de los Claveles, Gijo
´n, Asturias, Spain;
2
INRA and
3
ENVA, UMR 1198;
4
CNRS, FRE 2857,
Biologie du Developpement et Reproduction, Jouy-en-Josas, France
Contents
Because of the potential use of embryonic stem cells (ESC),
especially for genetic modifications, there is great interest in
establishing domestic animals-related ESCs. Unfortunately,
despite considerable efforts, validated ESC lines in species
other than mice and primates are yet to be isolated. In this
paper, we will summarize the current knowledge on bovine
ESCs in an attempt to understand why derivation of domestic
animal ESC is still unsuccessful and we will discuss some
promising future approaches.
Introduction
Embryonic stem cells (ESCs) were first isolated from
mouse embryos over 20 years ago (Evans and Kaufman
1981; Martin 1981). These cells, which are able to self-
renew indefinitely and to differentiate in vitro and in vivo
into derivatives of all three germ layers, were initially
used to study the differentiation process. Yet, soon after,
it became clear that ESCs provided an efficient route for
precise modification of the genome by gene targeting.
The first mutant mice derived from genetically engi-
neered ESCs were created, opening the doors to a new
era of animal transgenesis (Capecchi 1989).
The successful isolation and multiple applications of
mouse ESC (mESC) resulted in numerous efforts aimed
to establish ESCs in other species. Livestock production,
for example, would greatly benefit if farm animals ESCs
were established, in order to efficiently create genetically
modified farm animals bearing improved production
traits or increased disease resistances. Furthermore,
ESC lines from farm animals such as pigs, sheep or
cattle will enable to create transgenic animals for
modelling human diseases without some of the limita-
tions from the mouse model, i.e. short life span or a
physiology and anatomy very different from humans.
Unfortunately, although considerable effort has been
exerted to isolate and maintain ESC lines from domestic
animals, validated ESC lines in species other that mice
and primates are yet to be established.
Issues that need to be investigated are many, including
the identification of species-specific mechanisms under-
lying pluripotency and markers (for review, see Keefer
et al. 2007). A further hurdle is that most of the current
empirical approaches to obtain ESC from farm animals
closely followed procedures that were developed in
humans and mice. Nevertheless, data extrapolation from
these species has produced unsatisfactory results and
might have misled researchers from finding the func-
tional pathways that control pluripotency in ungulates.
In this paper, we analyse some of the currently
available data on bovine ESCs (bESC) to have a better
understanding of the failure to establish domestic
animals-validated ESC lines. We also describe some
new approaches that will perhaps enable the establish-
ment of these cell lines.
The Stage and the Source of Embryos
Embryonic stem cell lines have been successfully
isolated from mouse, monkey and human blastocysts,
although outstanding derivations have also been made
using embryos at pre-compaction stages (Eistetter 1988;
Delhaise et al. 1996; Strelchenko 1996; Mitalipova et al.
2001). Most attempts to isolate and culture bESCs have
been done with day 7–9 bovine blastocysts (Stice et al.
1996; Strelchenko 1996; Cibelli et al. 1998; Iwasaki
et al. 2000; Betts et al. 2001; Saito et al. 2003; Roach
et al. 2006; Mun
˜oz et al. 2008) although ESC-like cells
have also being isolated from day 12–14 embryos
(Gjørret and Maddox-Hyttel 2005). Yet, the optimal
timing of bovine pre-implantation development to
derive ESCs is still unknown.
Attempts to derive bESC from zygotes and early
cleavage stage embryos mostly failed (Strelchenko 1996;
Mitalipova et al. 2001), while only a single bovine
embryonic cell line, generated from a two-cell embryo,
has been cultured over 3 years (Mitalipova et al. 2001).
Yet, when bovine morulae were used as starting
material, efficient colony formation rates ranged over
60–70% (Stice et al. 1996; Strelchenko 1996). The
former author found that the embryonic stage (morulae
and Day-7 blastocysts) or source (in vivo and in vitro)
used did not influence the efficiency in establishing bESC
colonies. These results are nevertheless contrary to
studies reporting that Day-8 hatched blastocysts yield
a higher proportion of epiblast colonies than inner cell
masses (ICMs) isolated from Day-9 blastocysts (41%
and 13%, respectively) (Talbot et al. 1995). Although
bovine embryos have been used at different pre-implan-
tation stages to isolate bESC, the lack of standardized
and detailed protocols leads to an absence of homoge-
neous criteria to define efficiency of colony formation.
This hurdle precludes comparisons between published
data, making it impossible to conclude which develop-
mental stage entails the best starting material to isolate
ESCs. An alternative approach to answer this question
might be to extrapolate available data on mouse
embryonic development and mESC derivation; yet
Reprod Dom Anim 43 (Suppl. 4), 32–37 (2008); doi: 10.1111/j.1439-0531.2008.01229.x
ISSN 0936-6768
2008 The Authors. Journal compilation 2008 Blackwell Verlag
several species-specific differences must be carefully
considered prior to doing so. In mouse embryos, the
first differentiation event occurs at the late morula stage,
when the outer cells adopt an epithelial structure. This
event is followed by the formation of the blastocoel,
which marks the divergence of the first two lineages:
trophectoderm (TE) and inner cell mass. Upon blasto-
cyst expansion, differentiation continues with the ICM
forming two further lineages: the epiblast and the
primitive endoderm or hypoblast. Although epiblast,
hypoblast and TE lineages will be present in blastocysts
of all mammalian embryos, the time between fertiliza-
tion and formation is shorter in mouse than in other
species. Therefore, in order to isolate ESC from the
same type of cells used to derive mESC, i.e. epiblast cells
(Brook and Gardner 1997), we must consider differences
in the time scale of development and make sure that
embryonic stages equivalent to the murine counterparts
are used. In the pig blastocyst a defined epiblast is not
present before hatching, while in mice the epiblast is
already formed in blastocysts enclosed in the zona
pellucida. Therefore, derivation of ESCs from early
murine or porcine blastocysts is not equivalent. In
addition to differences in time scale of development,
recent data have reported important differences in
pathways that control pluripotency vs extra-embryonic
lineage restriction in mouse (Rossant 2007) and bovine
(Degrelle et al. 2005) (see below characterization of
bESC). Therefore any data extrapolation between
mouse and late-implanting species such as cattle should
be carefully considered.
Bovine embryos from different sources have been used
to isolate bESCs. Yet the only published experiment
aimed to compare the feasibility of in vitro- and in vivo-
derived embryos for the isolation of pluripotent cells
was undertaken by Talbot in 1995. Such a study
demonstrated that in vivo-derived blastocysts, especially
from early hatching blastocysts, were shown to be a
source of pluripotent epiblasts superior to their in vitro-
produced (IVP) counterparts.
The basis for any advantage by in vivo-produced
blastocyst to produce ESC lines is not known,
although a number of differences in morphology,
metabolic rates, gene expression and susceptibility to
cooling damage (Smith et al. 2005; Lonergan et al.
2006) have been reported between in vivo-derived and
IVP bovine embryos. It is possible that the reduced
number of cells present in the ICM of IVP bovine
embryos (Van Soom et al. 1996) might affect survival
of the ICM in culture, hindering the chances to
establish ESC lines from IVP embryos. In fact,
Anderson et al. (1994) assumed that the factor that
may affect survival of porcine ICMs in culture was the
number of cells of the ICMs.
In vivo-derived embryos might be a better source of
pluripotent cells, but their use as a starting material to
isolate ESC is expensive and laborious. Therefore, it
would be advisable to improve the procedures to derive
ESCs from IVP embryos, as well as the ability of IVP-
ICMs to yield ESCs. An obvious way to progress in this
aspect would be increasing numbers of cells in these
ICMs.
Characterization of Bovine ESC
Morphology, as well as the capacity to differentiate
in vitro through embryoid body (EB) formation, was one
of the two defining criteria initially used to identify
bESC cultures. Other traits such as small size, rounded
shape or high nucleus to cytoplasm ratio were used to
define bESC lines. Yet, cells belonging to TE and
visceral endoderm, which usually can be found in
blastocysts or isolated ICMs primary cultures, may be
confounded with ESC if solely morphological features
are used as evaluating criteria. Bovine blastocyst-derived
TE and endoderm cell lines have been thoroughly
characterized not only by morphological criteria but
also by the expression tissue-specific marker. For
instance, transferrin is a definitive marker for bovine
blastocyst-derived endoderm cell lines (Talbot et al.
2000). Therefore, the combined use of morphological
criteria and the analysis of extra-embryonic markers is
suggested to trul y identify bESC and or rule out the
presence of TE or visceral endoderm cells in ESC
cultures.
A useful strategy to characterize ESC lines is to
analyze the expression of pluripotency-related molecular
markers. Unfortunately, until now, no specific markers
have been identified in bovine. Therefore, markers
associated to pluripotency in other species (heterospec-
ific pluripotency markers) such as stage-specific embry-
onic antigens (SSEA-1, -3, -4) have been used to
characterize bESC. SSEAs are developmentally regu-
lated cell surface antigens expressed by murine and
human pluripotent cells (Fig. 1). mESCs strongly
express SSEA-1 (Solter and Knowles 1978; Gooi et al.
SSEA4
TRA-1-81
Bovine blastocyst Human blastocyst Murine blastocyst
SSEA1
SSEA4
TRA-1-81
TRA-1-60
SSEA4
TRA-1-81
TRA-1-60 SSEA1 SSEA1
SSEA1
TRA-1-60
Bovine ESC Murine ESCHuman ESC
SSEA4
TRA-1-81
SSEA4
TRA-1-81
SSEA1
TRA-1-60
TRA 1 81
TRA-1-60
Fig. 1. Expression of stage specific
embryonic antigens SSEA-1 and
SSEA-4 and keratin sulphate-ass-
ociated antigens TRA-1-60 and
TRA-1-81 in human and murine
blastocyst and embryonic stem
cells and in bovine blastocysts and
bovine ESC-like cells. Data from
human and mouse ICM and trop-
hectoderm are from Henderson
et al. (2002); data from human and
mouse embryonic stem cells are
from Boiani and Scho
¨ler (1997).
Data from bovine are from our lab
ESCs in Cattle 33
2008 The Authors. Journal compilation 2008 Blackwell Verlag
1981), whereas differentiated mESCs are characterized
by the loss of SSEA-1 expression and in some instances,
by the appearance of SSEA-3 and SSEA-4 (Solter and
Knowles 1979). In contrast, hESCs typically express
SSEA-3 and SSEA-4, but not SSEA-1 and their differ-
entiation is characterized by down-regulation of SSEA-3
and SSEA-4 and up-regulation of SSEA-1 (Andrews
et al. 1984; Fenderson et al. 1987). Undifferentiated
hESCs also express the keratin sulphate-associated
antigens TRA-1-60 and TRA-1-81 (Andrews et al.
1984). In bovine, a positive staining for SSEA-1,
SSEA-3 and SSEA-4 was reported in three embryonic
cell lines derived from pre-compaction embryos (Mita-
lipova et al. 2001). Similarly, SSEA-1 expression was
also detected by Saito et al. (2003), while none of the
bovine ES-like cells analysed by these authors were
found positive for SSEA-3 or SSEA-4. In contrast,
Wang et al. (2005) reported a positive SSEA-4 staining
in the absence of SSEA-1 staining in five ESC lines.
We have reported positive staining for SSEA-4, TRA-
1-60 and TRA-1-81 in bESC-like cells (Mun
˜oz et al.
2008). Unfortunately, the above antigens were not only
present in the ICM of bovine blastocysts but also in the
TE (Fig. 1). Therefore in bovine, these markers are not
specific for undifferentiated and or pluripotent cells.
The use of such markers to characterize bESC may
mislead researchers into isolating and culturing TE-
derived cells instead of ESCs.
The expression of SSEAs, in the TE of bovine
blastocysts, was unexpected, considering that for a long
time SSEAs have been used to characterize ‘undifferen-
tiated’ bESCs. Nevertheless, it was not totally surprising
as bovine TE cells show a slow differentiating phenotype
characterized by the co-expression of epiblast-specifying
genes (OCT-4, SOX-2, NANOG) and proteins (OCT-4,
NANOG) and trophoblast-specific genes (CDX-2,
HAND1, ETS-2, IFN-TAU, C12) (Kirchhof et al. 2000;
Degrelle et al. 2005; Mun
˜oz et al. 2008). Therefore in
bovine, the expression of markers which are associated
to pluripotency in other species (SSEA-4, TRA-1-60,
TRA-1-81, OCT-4, NANOG) is not restricted to plurip-
otent cells (Figs 1 and 2), which is a warning to validate
any pluripotency marker before its heterospecific use.
An additional difficulty to characterize bESC is that
available antibodies currently used to characterize ESCs
are produced using mouse or human proteins as
immunogens. Therefore, their ability to cross-react with
the appropriated bovine protein should be evaluated
before their use.
Cell Culture Conditions
Culture conditions close to those established for murine
ESC culture were successfully used to derive monkey
(Thomson et al. 1995) and human ESCs (Thomson
et al. 1998). Nevertheless, it soon became evident that
some factors required for the maintenance of mESC
pluripotency were not only dispensable in maintaining
hESC pluripotency but were also detrimental. As an
example, this occurred with BMP4, a member of the
transforming growth factor-b(TGF-b) family involved
in controlling mESC differentiation that induces differ-
entiation of human ESCs into trophoblast cells (Xu
et al. 2002). Since then, considerable amount of data
have been published over differences between mouse and
human pluripotency maintaining factors and signalling
pathways (for review see Renard et al. 2007; Fig. 3).
Until now, following a similar approach to primate ESC
isolation, most attempts to culture bESC have been
inspired by the original culture methods for mESC of
Evans and Kaufman (1981).
Bovine ESCs are usually cultured on mouse embry-
onic fibroblasts (primary MEF or transformed STO
cells). Culture media consists of Dulbecco’s Modified
Eagle’s Medium supplemented with foetal bovine serum,
L-glutamine, 2-bmercaptoethanol and different growth
factors, mostly leukaemia inhibitory factor (LIF) and
epidermal growth factor (EGF) (for comparison of some
bESC culture conditions see Table 1).
Yet it is likely that culture conditions suitable to
maintain mESC could be inadequate to maintain
undifferentiated bESC. Preliminary studies by Keefer
(2007) showed that the bovine ICM and its primary
outgrowths express the LIF receptor and gp130 trans-
ducer. Yet, LIF did not improve the establishment and
maintenance of ESCs from other ungulates (see Vack-
ova et al. 2007 for review) although its presence in pig
ESC culture medium prevented EB formation (Brevini
et al. 2007). It can be speculated that, such as in hESC,
stimulation of the STAT3 pathway by LIF might not
induce proliferation of ungulate ESCs. Similarly, some
growth factors found to suppress differentiation of
mESCs [such as TGB-b, EGF or insulin-like growth
factors (IGFs)] did not inhibit differentiation of porcine
Bovine blastocyst
Oct-4/OCT-4
Murine blastocyst
Human blastocyst
NANOG/NANOG
OCT-4/OCT-4
NANOG/NANOG
Nanog/NANOG
OCT-4/OCT-4
NANOG/NANOG
Murine ESCHuman ESC
N
Bovine ESC
NANOG
OCT-4
Nanog
Oct-4
NANOG
OCT-4
OCT-4/OCT-4 OCT-4/OCT-4
Fig. 2. Expression of oct-4 and
nanog mRNA and proteins in
bovine, human and murine
blastocyst and embryonic stem
cells. Data from bovine are from
Kirchhof et al. (2000), Degrelle
et al. (2005), Gjørret and Maddox-
Hyttel (2005), Wang et al. (2005)
and Mun
˜oz et al. (2008). Data
from human are from Adjaye et al.
(2005), Cauffman et al. (2005),
Chambers et al. (2003) and Kimber
et al. (2008). Data from mouse are
from Palmieri et al. (1994), Kirch-
hof et al. (2000), Chambers et al.
(2003) and Hatano et al. (2005)
34 M Mun
˜oz, C Dı
´ez, JN Caaman
˜o, A Jouneau, I Hue and E Go
´mez
2008 The Authors. Journal compilation 2008 Blackwell Verlag
stem cells (Hochereau-de Reviers and Perreau 1993;
Prelle et al. 1994).
The inability of currently used culture conditions to
meet the nutrient or growth factor requirements of bESC
is one of the possible explanations for the failure to
isolate these cells. Identification of specific pluripotency
signalling pathways will help to determine which growth
factors are beneficial or which ones are inappropriate for
establishing a successful bESC culture.
In Vitro and In Vivo Differentiation of bESC
The differentiation ability of bESC in vitro is evaluated
by EB formation. EBs are aggregates of stem cells whose
development is reminiscent of early embryogenesis.
Maintenance of bESC in a suspension culture (Strel-
chenko 1996) or in the absence of a feeder layer (Saito
et al. 2003; Wang et al. 2005) initiates the formation of
EBs. The EBs are composed of two layers of cells,
ectoderm-like cells covered by a thin layer of endoderm
like cells, with heterogeneous cellular particles within the
cavity. The cells of bovine EBs give rise to a wide variety
of differentiated cell types, including derivatives of the
three germ layers (Saito et al. 2003; Wang et al. 2005).
This ability is a proof of their pluripotent-differentiation
character in vitro.
The ability of bESC to participate in the embryogen-
esis has been proven only after a short propagation
period in vitro. Chimeric transgenic calves have been
born after injection of bESC (passage 3) into cleavage
stage embryos (Cibelli et al. 1998), embryo aggregation
with bESC (passage 9–13) (Iwasaki et al. 2000). Yet
integration of ESCs into the germ line, one of the
properties used to define ESCs, have not been achieved
in any of these experiments.
Calves were also successfully cloned using ES-like
cells as donor nuclei (short cultured ICMs or passage
14–18) (Sims and First 1994; Saito et al. 2003), but the
use of ESC in nuclear transfer can not be taken as a
proof of pluripotency as cloned animals have been
produced from fully differentiated somatic cell nuclei
(Kato et al. 2000; Wakayama and Yanagimachi 2001).
Conclusions
The above summarized data indicate that although it
is possible to isolate bESC, suitable conditions for
BMP4 LIF
Mouse/Human
Activin/Nodal TGFβFGF
2
BMP4 LIF
Smad 1/5/8 STAT3/5 Smad 2/3 MAPK/ERK STAT3
Pluripotency Pluripotency Pluripotency Differentation
Ungulates
FGF
2
LIF
No effects on ESCPositive effects on No
establishment/maintenance ESC proliferation
Human
Mouse
Fig. 3. Pluripotency and differen-
tiation mediating pathways in
murine, human and embryonic
stem cells. Data from murine and
human are from Renard et al.
(2007). Data from ungulates were
reviewed by Vackova et al. (2007)
Table 1. A selection of reports on the culture media employed to
derive bovine embryonic stem cells
Reference Feeder layer Cell culture media
Talbot et al. (1995) Murine STO cells DMEM-M 199
FBS
b-ME
L-Glutamine
Non-essential
amino acids
Nucleosides
Mitalipova et al. (2001) Murine embryonic
fibroblasts
Alpha-MEM
FBS
b-ME
L-Glutamine
Saito et al. (2003) Murine STO cells MEM
FBS
b-ME
hLIF
hEGF
Wang et al. (2005) Murine embryonic
fibroblasts
Knock-out DMEM
FBS
b-ME
L-Glutamine
Non-essential
amino acids
hLIF
bFGF
Gjørret and Maddox-Hyttel
(2005)
Murine STO cells Knock-out DMEM
FCS
b-ME
L-Glutamine
MEM amino acids
hLIF
bFGF
Mun
˜oz et al. (2008) Bovine embryonic
fibroblasts
DMEM
FCS
MEM non-essential
amino acids
bFGF
bFGF, basic fibroblast growth factor; b-ME, b-mercaptoethanol; DMEM,
Dulbecco’s Modified Eagle’s Medium; FBS, foetal bovine serum; FCS, foetal
calf serum; hEGF, human epidermal growth factor; hLIF, human leukaemia
inhibitory factor; MEM, Minimum Essential Medium.
ESCs in Cattle 35
2008 The Authors. Journal compilation 2008 Blackwell Verlag
preventing spontaneous differentiation and senescence of
these cells are yet to be established. Similarly, validated
ESC lines have not been produced yet in other domestic
species (for review see Keefer et al. 2007).
Possible explanations for this failure are many,
including inadequate starting material or misleading
characterization of cell lines because of the inadequate
use of heterospecific markers. It is likely that the
inability to optimize culture conditions might have been
the biggest obstacle precluding success.
Until now empirical approaches, based almost exclu-
sively on protocols developed for derivation of mouse
stem cells, have been used to design cell culture environ-
ments for ES derivation in other species. Nevertheless,
taking in account the increasing differences reported in
the mechanisms and factors that regulate pluripotency
and self-renewal among mammals (for review see Renard
et al. 2007), it has become clear that alternative and or
complementary strategies will have to be applied.
Several techniques are currently used to identify the
gene expression profile of ESCs. These techniques include
serial analysis of gene expression, subtractive hybridiza-
tion, representational difference analysis, cDNA micro-
arrays and oligonucleotide microarray technologies. The
use of the above tools have allowed to identify unique
expression patterns of transcripts found in mouse and
human stem cells in comparison with differentiated
counterparts. The genes and pathways that appear to be
related to stemness in mouse and human ESCs are good
candidates to analyse basic mechanism governing pluri-
potency in mammalian ESCs. Furthermore, future tran-
scriptomic analysis among ESCs from different species
will help to identify species-specific pluripotency related
genes and pathways which could be used to design
tailored cell culture environments that enable ESC
derivation from other species.
Nuclear transfer could also provide an alternative
approach to improve ESC derivation (Renard et al.
2007). Studies with different strains of mice have shown
that the isolation of ESC can be hampered by the use of
different genotypes (Kawase et al. 1994). Thus in species
where the genetic background variability is high, the use
of genetically identical ICMs isolated from blastocysts
obtained by nuclear transfer could simplify the screening
for cell culture environments that would yield stable
ESC lines.
All the data gathered actually so far and that may be
collected in the near future about ESCs will help unravel
the mystery of the pluripotent state enabling the
establishment of ESCs from domestic animals.
Acknowledgements
Spanish Ministry of Science and Education (Project AGL2005–04479).
Dr M. Mun
˜oz is supported by a grant from FICYT.
Conflicts of interest
Authors of this paper have no conflicts of interest.
Author Contributions
All authors have contributed equally to write this paper.
References
Adjaye J, Huntress J, Herwig R, BenKahla A, Brink TC,
Wierling C, Hultschig C, Groth D, Yaspo ML, Picton HM,
Gosden R, Lehrach H, 2005: Primary differentiation in the
human blastocyst: comparative molecular portraits of inner
cell mass and trophectoderm cells. Stem Cells 23, 1514–1525.
Anderson GB, Choi SJ, Bondurant RH, 1994: Survival of
porcine inner cell masses in culture after injection into
blastocysts. Theriogenology 42, 204–212.
Andrews PW, Damjanov I, Simon D, Banting GS, Carlin C,
Dracopoli NC, Fogh J, 1984: Pluripotency embryonal
carcinoma clones derived from human teratocarcinoma cell
line Tera-2: differentiation in vivo and in vitro. Lab Invest 50,
147–162.
Betts D, Bordignon V, Hill J, Winger Q, Westhusin M, Smith L,
King W, 2001: Reprogramming of telomerase activity and
rebuilding of telomere length in cloned cattle. Proc Natl Acad
Sci U S A 98, 1077–1082.
Boiani M, Scho
¨ler HR, 1997: Regulatory networks in embryo-
derived pluripotent stem cells. Nat Rev Mol Cell Biol 6,
872–884.
Brevini TA, Antonini S, Cillo F, Crestan M, Gandolfi F, 2007:
Porcine embryonic stem cells: facts, challenges and hopes.
Theriogenology 68, S206–S213.
Brook FA, Gardner RL, 1997: The origin and efficient
derivation of embryonic stem cells in the mouse. Proc Natl
Acad Sci U S A 94, 5709–5712.
Capecchi MR, 1989: Altering the genome by homologous
recombination. Science 244, 1288–1292.
Cauffman G, Van de Velde H, Liebaers I, Steirteghem A, 2005:
Oct-4 mRNA and protein expression during human preim-
plantation development. Mol Hum Reprod 11, 173–181.
Chambers I, Colby D, Robertson M, Nichols J, Lee S,
Tweedie S, Smith A, 2003: Functional expression cloning of
Nanog, a pluripotency sustaining factor in embryonic stem
cells. Cell 113, 643–655.
Cibelli JB, Stice SL, Golueke PJ, Kane JJ, Jerry J, Blackwell C,
Ponce de Leo
´n FA, Robl JM, 1998: Transgenic bovine
chimeric offspring produced from somatic cell-derived stem-
like cells. Nat Biotechnol 16, 642–646.
Degrelle SA, Campion E, Cabau C, Piumi F, Reinaud P,
Richard C, Renard JP, Hue E, 2005: Molecular evidence for
a critical period in mural trophoblast development in bovine
blastocysts. Dev Biol 288, 448–460.
Delhaise F, Bralion V, Schuurbiers N, Dessy F, 1996:
Establishment of an embryonic stem cell line from 8-cell
stage mouse embryos. Eur J Morphol 34, 237–243.
Eistetter HR, 1988: A mouse pluripotent embryonal stem cell
line stage-specifically regulates expression of homeo-box
containing DNA sequences during differentiation in vitro.
Eur J Cell Biol 45, 315–321.
Evans MJ, Kaufman MH, 1981: Establishment in culture
of pluripotent cells from mouse embryo. Nature 292, 154–
156.
Fenderson BA, Andrews PW, Nudelman E, Clausen H,
Hakamori S, 1987: Glycolipid core structure switching from
globo- to lacto- and gangli-series during retinoic acid-
induced differentiation of TERA-2-derived human embryo-
nal carcinoma cells. Dev Biol 122, 21–34.
Gjørret JO, Maddox-Hyttel P, 2005: Attempts towards deri-
vation and establishment of bovine embryonic stem cell-like
cultures. Reprod Fertil Dev 17, 113–124.
Gooi HC, Feizi T, Kapadia A, Knowles BB, Solter D, Evans
MJ, 1981: Stage-specific embryonic antigen involves alpha 1
goes to 3 fucosylated type 2 blood group chains. Nature 292,
156–158.
Hatano SY, Tada M, Kimura H, Yamaguchi S, Kono T,
Nakano T, Suemori H, Nakatsuji N, Tada T, 2005:
36 M Mun
˜oz, C Dı
´ez, JN Caaman
˜o, A Jouneau, I Hue and E Go
´mez
2008 The Authors. Journal compilation 2008 Blackwell Verlag
Pluripotential competence of cells associated with Nanog
activity. Mech Dev 122, 67–79.
Henderson JK, Draper JS, Baillie HS, Fishel S, Thomson JA,
Moore H, Andrews PW, 2002: Preimplantation human
embryos and embryonic stem cells show comparable
expression of stage-specific embryonic antigens. Stem Cells
20, 329–337.
Hochereau-de Reviers MT, Perreau C, 1993: In vitro culture of
embryonic disc cells from porcine blastocysts. Reprod Nutr
Dev 33, 475–483.
Iwasaki S, Campbell KH, Galli C, Akiyama K, 2000:
Production of live calves derived from embryonic stem-like
cells aggregated with tetraploid embryos. Biol Reprod 62,
470–475.
Kato Y, Tani T, Tsunoda Y, 2000: Cloning of calves from
various somatic cell types of male and female adult,
newborn and fetal cows. J Reprod Fertil 20, 231–237.
Kawase E, Suemori H, Takahashi N, Okazaki K, Hashimoto K,
Nakatsuji N, 1994: Strain difference in establishment of
mouse embryonic stem (ES) cell lines. Int J Dev Biol 38, 385–
390.
Keefer CL, Pant D, Blomberg L, Talbot NC, 2007: Challenges
and prospects for the establishment of embryonic stem cell
lines of domestic ungulates. Anim Reprod Sci 98, 147–168.
Kimber SJ, Sneddon SF, Bloor DJ, El-Bareg AM, Hawkhead
JA, Metcalfe AD, Houghton FD, Leese HJ, Rutherford A,
Lieberman BA, Brison DR, 2008: Expression of genes
involved in early cell fate decisions in human embryos and
their regulation by growth factors. Reproduction 135, 635–
647.
Kirchhof N, Carnwath JW, Lemme E, Anastassiadis K,
Scho
¨le H, Niemann H, 2000: Expression pattern of Oct-4
in preimplantation embryos of different species. Biol
Reprod 63, 1698–1705.
Lonergan P, Fair T, Corcoran D, Evans AC, 2006: Effect of
culture environment on gene expression and developmental
characteristics in IVF-derived embryos. Theriogenology 65,
137–152.
Martin GR, 1981: Isolation of a pluripotent cell line from early
mouse embryos cultured in medium conditioned by terato-
carcinoma stem cells. Proc Natl Acad Sci U S A 78, 7634–
7638.
Mitalipova M, Beyhan Z, First N, 2001: Pluripotency of
bovine embryonic cell line derived from precompacting
embryos. Cloning 3, 59–67.
Mun
˜oz M, Rodriguez A, De Frutos C, Caaman
˜o JN, Dı
´ez C,
Facal N, Go
´mez E, 2008: Conventional pluripotency mark-
ers are unspecific for bovine embryonic derived cell-lines.
Theriogenology 69, 1159–1164.
Palmieri SL, Peter W, Hess H, Scho
¨ler HR, 1994: Oct-4
transcription factor is differentially expressed in the mouse
embryo during establishment of the first two extraembryonic
cell lineages involved in implantation. Dev Biol 166, 259–
267.
Prelle K, Wobus AM, Wolf E, Neubert N, Holtz W, 1994:
Effects of growth factors on the in vitro development of
porcine inner cell masses isolated by calcium ionophore.
J Reprod Fertil 13, 41.
Renard JP, Maruotti J, Jouneau A, Vignon X, 2007: Nuclear
reprogramming and pluripotency of embryonic cells: Appli-
cation to the isolation of embryonic stem cells in farm
animals. Theriogenology 68, S196–S205.
Roach M, Wang L, Yang X, Tian XC, 2006: Bovine
embryonic stem cells. Methods Enzymol 418, 21–37.
Rossant J, 2007: Stem cells and lineage development in the
mammalian blastocyst. Reprod Fertil Dev 19, 111–118.
Saito S, Sawai K, Ugai H, Moriyasu S, Minamihashi A,
Yamammoto Y, Hirayama H, Kageyama S, Pan J, Murata T,
Kobayashi Y, Obata Y, Yokoyama KK, 2003: Generation of
cloned calves and transgenic chimeric embryos from bovine
embryonic stem-like cells. Biochem Biophys Res Commun
309, 104–113.
Sims M, First NLM, 1994: Production of calves by transfer of
nuclei from cultured inner cell mass cells. Proc Natl Acad
Sci U S A 91, 6143–6147.
Smith SL, Everts RE, Tian XC, Du F, Sung LY, Rodriguez-
Zas SL, Jeong BS, Renard JP, Lewin HA, Yang X, 2005:
Global gene expression profiles reveal significant nuclear
reprogramming by the blastocyst stage after cloning. Proc
Natl Acad Sci U S A 102, 17582–17587.
Solter D, Knowles BB, 1978: Monoclonal antibody defining a
stage-specific mouse embryonic antigen (SSEA-1). Proc Natl
Acad Sci U S A 75, 5565–5569.
Solter D, Knowles BB, 1979: Developmental stage-specific
antigens during mouse embryogenesis. Curr Top Dev Biol
13, 139–165.
Stice SL, Strelchenko N, Keefer CL, Matthews L, 1996:
Pluripotent bovine embryonic cell lines diret embryonic
development following nuclear transfer. Biol Reprod 54,
100–110.
Strelchenko N, 1996: Bovine pluripotent stem cells. Theriog-
enology 45, 131–140.
Talbot NC, Powell AM, Rexroad CE Jr, 1995: In vitro
pluripotency of epiblasts derived from bovine blastocysts.
Mol Reprod Dev 42, 35–52.
Talbot NC, Caperna TJ, Edwards JL, Garrett W, Wells KD,
Ealy AD, 2000: Bovine blastocyst-derived trophectoderm
and endoderm cell cultures: interferon tau and transferrin
expression as respective in vitro markers. Biol Reprod 62,
235–247.
Thomson JA, Kalishman J, Golos TG, Durning M, Harris CP,
Becker RA, Hearn JP, 1995: Isolation of a primate
embryonic stem cell line. Proc Natl Acad Sci U S A 92,
7844–7848.
Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA,
Swiergiel JJ, Marshall VS, Jones JM, 1998: Embryonic stem
cell lines derived from human blastocysts. Science 282,
1145–1147. Erratum in: Science 1998 282, 1827.
Vackova I, Ungrova A, Lopes F, 2007: Putative embryonic
stem cell lines from pig embryos. J Reprod Dev 53, 1137–
1149.
Van Soom A, Boerjan M, Ysebaert MT, De Kruif A, 1996:
Cell allocation to the inner cell mass and the trophectoderm
in bovine embryos cultured in two different media. Mol
Reprod Dev 45, 171–182.
Wakayama T, Yanagimachi R, 2001: Mouse cloning with
nucleus donor cells of different age and type. Mol Reprod
Dev 58, 376–383.
Wang L, Duan E, Sung L, Jeong B, Yang X, Tian C, 2005:
Generation and characterization of pluripotent stem cells
from cloned bovine embryos. Biol Reprod 73, 149–155.
Xu RH, Chen X, Li DS, Li R, Addicks GC, Glennon C,
Zwaka TP, Thomson JA, 2002: BMP4 initiates human
embryonic stem cell differentiation to trophoblast. Nat
Biotechnol 20, 1261–1264.
Submitted: 18 May 2008
Author’s address (for correspondence): Marta Mun
˜oz, Area de
Gene
´tica y Reproduccio
´n, SERIDA, Camino de los Claveles 604,
33203 Gijo
´n, Asturias, Spain. E-mail: mmunoz@serida.org
ESCs in Cattle 37
2008 The Authors. Journal compilation 2008 Blackwell Verlag
... They are glycoprotein and generally have a molecular weight between 60-80kDa and pI 3-6 [42]. Due to broad substrate specificity as well as wide reaction capabilities, laccase possess promising applications in textile dye bleaching [51], pulp bleaching [84], bioremediation of soils as well as water [65,120] and development of biosensors and biofuel cells [115,114,109]. ...
... Moreover, they also express higher levels of cancerous proteins, cytokines and adhesion molecules which move to myeloma cells after modulating tumor growth in vivo [46]. Exosomes from BM-MSCs increase the expression level of vascular endothelial growth factor (VEGF) and activate the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway to promote tumor growth [65]. Mast cell derived exosomes stimulate the KIT stem cell factor (SCF) signaling pathway which promote tumor growth in recipient lung adenocarcinoma cells [60]. ...
... Studies also explain that PDGF-BB induces proliferation as well as migration in MSCs [31]. Moreover, PDGFR-β inhibits osteogenesis while PDGFR-α induces osteogenesis [65]. Akt signaling mediates induction and suppression of osteogenesis by PDGFR signaling [23]. ...
Chapter
Nanotechnology and the use of nanomaterials is a new area of science and is emerging rapidly day by day. Due to increased surface area, reactivity and advanced characteristics nanoparticles have many applications in diverse fields. Varieties of nanoparticles are manufactured like fullerenns, carbon nanotube, metals, metals oxides, q-dots, etc. These nanoparticles enter into the environment through waste products and cause negative effects on flora and fauna of the ecosystem. Due to small size and high reactivity, nanoparticles can enter into living organisms also and cause oxidative stress and inflammatory reactions. Number of studies have been carried out to manifest ecotoxicity of nanoparticles and were found to be highly toxic to the organisms. Therefore it is urgent need to undergo detailed research on the ecotoxicity of nanoparticles and to develop methods to remediate them from the environment. Otherwise situation may become worse in the future.
... In other species, however, these criteria were not yet reproducible until the advent of iPSC technology. In cattle, for example, isolation of embryonic cells results in colonies that may correspond to some of the criteria required for human and murine cells; however, in vitro culture of these stem cells revealed differences from the characteristics outlined above in several reports [37][38][39][40][41]. ...
Article
Full-text available
Abstract Background: Induced pluripotent stem cells (iPSCs) have enormous potential in developmental biology studies and in cellular therapies. Although extensively studied and characterized in human and murine models, iPSCs from animals other than mice lack reproducible results. Methods: Herein, we describe the generation of robust iPSCs from equine and bovine cells through lentiviral transduction of murine or human transcription factors Oct4, Sox2, Klf4, and c-Myc and from human and murine cells using similar protocols, even when different supplementations were used. The iPSCs were analyzed regarding morphology, gene and protein expression of pluripotency factors, alkaline phosphatase detection, and spontaneous and induced differentiation. Results: Although embryonic-derived stem cells are yet not well characterized in domestic animals, generation of iPS cells from these species is possible through similar protocols used for mouse or human cells, enabling the use of pluripotent cells from large animals for basic or applied purposes. Herein, we also infer that bovine iPS (biPSCs) exhibit similarity to mouse iPSCs (miPSCs), whereas equine iPSs (eiPSCs) to human (hiPSCs). Conclusions: The generation of reproducible protocols in different animal species will provide an informative tool for producing in vitro autologous pluripotent cells from domestic animals. These cells will create new opportunities in animal breeding through transgenic technology and will support a new era of translational medicine with large animal models. Keywords: Bovine, Cellular reprogramming, Equine, Induced pluripotency, iPSCs, stem cells
... Although ESC cultures have been established for mice more than three decades ago (Evans & Kaufman, 1981), attempts to establish cultures of authentic ESCs or epiblast stem cells in livestock species have been largely unsuccessful (Ezashi et al., 2016;Munoz et al., 2008). ...
Article
Full-text available
Mechanisms that direct reprogramming of differentiated somatic cells to induced pluripotent stem cells (iPSCs), albeit incomplete in understanding, are highly conserved across all mammalian species studied. Equally, proof of principle that iPSCs can be derived from domestic cattle has been reported in several publications. In our efforts to derive and study bovine iPSCs, we encountered inadequacy of methods to generate, sustain, and characterize these cells. Our results suggest that iPSC protocols optimized for mouse and human somatic cells do not effectively translate to bovine somatic cells, which show some refractoriness to reprogramming that also affects sustenance. Moreover, methods that enhance reprogramming efficiency in mouse and human cells had no effect on improving bovine cell reprogramming. Although use of retroviral vectors coding for bovine OCT4, SOX2, KLF4, cMYC, and NANOG appeared to produce consistent iPSC‐like cells from both fibroblasts and cells from the Wharton's jelly, these colonies could not be sustained. Use of bovine genes could successfully reprogram both mouse and human cells. These findings indicated either incomplete reprogramming and/or discordant/inadequate culture conditions for bovine pluripotent stem cells. Therefore, additional studies that advance core knowledge of bovine pluripotency are necessary before any anticipated iPSC‐driven bovine technologies can be realized.
... There also have been attempts to generate ESCs in domestic animals [3][4][5][6]. In agricultural perspective, the establishment of ESCs in domestic ungulates is able to provide a more efficient way to produce genetically-modified animals. ...
Article
Full-text available
Although there are many studies about pluripotent stem cells, little is known about pluripotent pathways and the difficulties of maintaining the pluripotency of bovine cells in vitro. Here, we investigated differently expressed genes (DEG) in bovine embryo-derived stem-like cells (eSLCs) from various origins to validate their distinct characteristics of pluripotency and differentiation. We identified core pluripotency markers and additional markers which were not determined as pluripotency markers yet in bovine eSLCs. Using the KEGG database, TGFβ, WNT, and LIF signaling were related to the maintenance of pluripotency. In contrast, some DEGs related to the LIF pathway were down-regulated, suggesting that reactivation of the pathway may be required for the establishment of true bovine embryonic stem cells (ESCs). Interestingly, oncogenes were co-down-regulated, while tumor suppressor genes were co-up-regulated in eSLCs, implying that this pattern may induce abnormal teratomas. These data analyses of signaling pathways provide essential information on authentic ESCs in addition to providing evidence for pluripotency in bovine eSLCs.
Article
Full-text available
The recent progress in derivation of pluripotent stem cells (PSCs) from farm animals opens new approaches not only for reproduction, genetic engineering, treatment and conservation of these species, but also for screening novel drugs for their efficacy and toxicity, and modelling of human diseases. Initial attempts to derive PSCs from the inner cell mass of blastocyst stages in farm animals were largely unsuccessful as either the cells survived for only a few passages, or lost their cellular potency; indicating that the protocols which allowed the derivation of murine or human embryonic stem (ES) cells were not sufficient to support the maintenance of ES cells from farm animals. This scenario changed by the innovation of induced pluripotency and by the development of the 3 inhibitor culture conditions to support naïve pluripotency in ES cells from livestock species. However, the long-term culture of livestock PSCs while maintaining the full pluripotency is still challenging, and requires further refinements. Here, we review the current achievements in the derivation of PSCs from farm animals, and discuss the potential application areas.
Article
Full-text available
Efficient isolation of embryonic stem (ES) cells from pre-implantation porcine embryos has remained a challenge. Here, we describe the derivation of porcine embryonic stem-like cells (pESLCs) by seeding the isolated inner cell mass (ICM) from in vitro-produced porcine blastocyst into α-MEM with basic fibroblast growth factor (bFGF). The pESL cells kept the normal karyotype and displayed flatten clones, similar in phenotype to human embryonic stem cells (hES cells) and rodent epiblast stem cells. These cells exhibited alkaline phosphatase (AP) activity and expressed pluripotency markers such as OCT4, NANOG, SOX2, SSEA-4, TRA-1-60, and TRA-1-81 as determined by both immunofluorescence and RT-PCR. Additionally, these cells formed embryoid body (EB), teratomas and also differentiated into 3 germ layers in vitro and in vivo. Microarray analysis showed the expression of the pluripotency markers, PODXL, REX1, SOX2, KLF5 and NR6A1, was significantly higher compared with porcine embryonic fibroblasts (PEF), but expression of OCT4, TBX3, REX1, LIN28A and DPPA5, was lower compared to the whole blastocysts or ICM of blastocyst. Our results showed that porcine embryonic stem-like cells can be established from in vitro-produced blastocyst-stage embryos, which promote porcine naive ES cells to be established.
Article
In this study, we screened the possible differentially expressed genes in bovine Embryonic Stem Cell (ESC)-like cells with different clone morphologies and passages via Differential Display Reverse Transcription PCR (DDRT-PCR) technique in combination with sequence analysis and GenBank BLAST. The results showed that among the bovine ESC-like cells with different clone morphologies, six differentially expressed fragments were identified highly homological with six known genes, namely: RPL9, LOC100850994, AMP, EPL31, Erbb2ip and CLIP I. Real-time quantitative PCR (qRT-PCR) results revealed that the expression levels of RPL9, LOC100850994 andRPL31 genes were significantly different among the bovine ESC-like cells with slate, block and bubble clone morphologies and Western blotting data showed that the expression levels of RPL9 were significantly different among the cells with the three clone morphologies. From the bovine ESC-like cells of different passages, seven differentially expressed fragments were identified highly homological with seven other known genes, namely: IK, TKDP1, BZW, PRL9, BPL31, PP42 and IGBP1. qRT-PCR results revealed that the expression levels of IK, TKDP1, BZW, PRL9, RP42 and IGBP1 were significantly different among the bovine ESC-like cells of 1 st and 10th passages. Western blotting data showed that protein expression levels of all these genes were significantly higher in passage 1 bovine ESC-like cells than passage 10 cells.
Article
The establishment of porcine embryonic stem cells (ESCs) would have great impact in biomedical studies and preclinical trials through their use in genetic engineering. However, authentic porcine ESCs have not been established until now. In this study, a total of seven putative ESC lines were derived from porcine embryos of various origins, including in vitro fertilization, parthenogenetic activation, and, in particular, induced pluripotent stem (iPS) nuclear transfer (NT) from a donor cell with induced pluripotent stem cells (iPSCs). To characterize these cell lines, several assays including an assessment of intensive alkaline phosphatase activity, karyotyping, embryoid body formation, expression analysis of the pluripotency-associated markers, and the three germ layerassociated markers were performed. Based on quantitative polymerase chain reaction, the expression levels of REX1 and FGFR2 in iPS-NT lines were higher than those of cells of other origins. Additionally, only iPS-NT lines showed multiple aberrant patterns of nuclear foci elucidated by immunofluorescence staining of H3K27me3 as a marker of the state of X chromosome inactivation and a less mature form of mitochondria like naive ESCs, by transmission electron microscopy. Together, these data suggested that established putative porcine ESC lines generally exhibited a primed pluripotent state, like human ESCs. However, iPS-NT lines have especially unique characteristics distinct from other origins because they have more epigenetic instability and naive-like mitochondrial morphology than other putative ESC lines. This is the first study to establish and characterize the iPSC-derived putative ESC lines and compare them with other lines derived from different origins in pigs.
Article
Full-text available
We established and maintained somatic cell nuclear transfer embryo-derived stem-like cells (SCNT-eSLCs) from the traditional Korean beef cattle species, HanWoo (Bos taurus coreanae). Each SCNT blastocyst was placed individually on a feeder layer with culture medium containing three inhibitors of differentiation (3i). Primary colonies formed after 2-3 days of culture and the intact colonies were passaged every 5-6 days. The cells in each colony showed embryonic stem cell-like morphologies with a distinct boundary and were positive to alkaline phosphatase staining. Immunofluorescence and reverse transcription-polymerase chain reaction analyses also confirmed that these colonies expressed pluripotent markers. The colonies were maintained over 50 passages for more than 270 days. The cells showed normal karyotypes consisting of 60 chromosomes at Passage 50. Embryoid bodies were formed by suspension culture to analyse in vitro differentiation capability. Marker genes representing the differentiation into three germ layers were expressed. Typical embryonal carcinoma was generated after injecting cells under the testis capsule of nude mice, suggesting that the cultured cells may also have the potential of in vivo differentiation. In conclusion, we generated eSLCs from SCNT bovine embryos, using a 3i system that sustained stemness, normal karyotype and pluripotency, which was confirmed by in vitro and in vivo differentiation.
Article
Full-text available
Nuclear reprogramming requires the removal of epigenetic modifications imposed on the chromatin during cellular differentiation and division. The mammalian oocyte can reverse these alterations to a state of totipotency, allowing the production of viable cloned offspring from somatic cell nuclei. To determine whether nuclear reprogramming is complete in cloned animals, we assessed the telomerase activity and telomere length status in cloned embryos, fetuses, and newborn offspring derived from somatic cell nuclear transfer. In this report, we show that telomerase activity was significantly (P < 0.05) diminished in bovine fibroblast donor cells compared with embryonic stem-like cells, and surprisingly was 16-fold higher in fetal fibroblasts compared with adult fibroblasts (P < 0.05). Cell passaging and culture periods under serum starvation conditions significantly decreased telomerase activity by approximately 30–50% compared with nontreated early passage cells (P < 0.05). Telomere shortening was observed during in vitro culture of bovine fetal fibroblasts and in very late passages of embryonic stem-like cells. Reprogramming of telomerase activity was apparent by the blastocyst stage of postcloning embryonic development, and telomere lengths were longer (15–23 kb) in cloned fetuses and offspring than the relatively short mean terminal restriction fragment lengths (14–18 kb) observed in adult donor cells. Overall, telomere lengths of cloned fetuses and newborn calves (≈20 kb) were not significantly different from those of age-matched control animals (P > 0.05). These results demonstrate that cloned embryos inherit genomic modifications acquired during the donor nuclei's in vivo and in vitro period but are subsequently reversed during development of the cloned animal.
Article
Full-text available
Distinctive properties of stem cells are not autonomously achieved, and recent evidence points to a level of external control from the microenvironment. Here, we demonstrate that self-renewal and pluripotent properties of human embryonic stem (ES) cells depend on a dynamic interplay between human ES cells and autologously derived human ES cell fibroblast-like cells (hdFs). Human ES cells and hdFs are uniquely defined by insulin-like growth factor (IGF)- and fibroblast growth factor (FGF)-dependence. IGF 1 receptor (IGF1R) expression was exclusive to the human ES cells, whereas FGF receptor 1 (FGFR1) expression was restricted to surrounding hdFs. Blocking the IGF-II/IGF1R pathway reduced survival and clonogenicity of human ES cells, whereas inhibition of the FGF pathway indirectly caused differentiation. IGF-II is expressed by hdFs in response to FGF, and alone was sufficient in maintaining human ES cell cultures. Our study demonstrates a direct role of the IGF-II/IGF1R axis on human ES cell physiology and establishes that hdFs produced by human ES cells themselves define the stem cell niche of pluripotent human stem cells.
Article
This report describes the establishment directly from normal preimplantation mouse embryos of a cell line that forms teratocarcinomas when injected into mice. The pluripotency of these embryonic stem cells was demonstrated conclusively by the observation that subclonal cultures, derived from isolated single cells, can differentiate into a wide variety of cell types. Such embryonic stem cells were isolated from inner cell masses of late blastocysts cultured in medium conditioned by an established teratocarcinoma stem cell line. This suggests that such conditioned medium might contain a growth factor that stimulates the proliferation or inhibits the differentiation of normal pluripotent embryonic cells, or both. This method of obtaining embryonic stem cells makes feasible the isolation of pluripotent cells lines from various types of noninbred embryo, including those carrying mutant genes. The availability of such cell lines should made possible new approaches to the study of early mammalian development.
Article
During gastrulation, a cascade of inductive tissue interactions converts pre-existing polarity in the mammalian embryo into antero-posterior pattern. This process is triggered by Nodal, a protein related to transforming growth factor-beta (TFG-beta) that is expressed in the epiblast and visceral endoderm, and its co-receptor Cripto, which is induced downstream of Nodal. Here we show that the proprotein convertases Spc1 and Spc4 (also known as Furin and Pace4, respectively) are expressed in adjacent extraembryonic ectoderm. They stimulate Nodal maturation after its secretion and are required in vivo for Nodal signalling. Embryo explants deprived of extraembryonic ectoderm phenocopy Spc1(-/-); Spc4(-/-) double mutants in that endogenous Nodal fails to induce Cripto. But recombinant mature Nodal, unlike uncleaved precursor, can efficiently rescue Cripto expression. Cripto is also expressed in explants treated with bone morphogenetic protein 4 (BMP4). This indicates that Nodal may induce Cripto through both a signalling pathway in the embryo and induction of Bmp4 in the extraembryonic ectoderm. A lack of Spc1 and Spc4 affects both pathways because these proteases also stimulate induction of Bmp4.
Article
Twenty-four calves were cloned from six somatic cell types of female and male adult, newborn and fetal cows. The clones were derived from female cumulus (n = 3), oviduct (n = 2) and uterine (n = 2) cells, female and male skin cells (n = 10), and male ear (n = 5) and liver (n = 2) cells. On the basis of the number of cloned embryos transferred (n = 172) to surrogate cows, the overall rate of success was 14%, but based on the number of surrogate mothers that became pregnant (n = 50), the success rate was 48%. Cell nuclei from uterus, ear and liver cells, which have not been tested previously, developed into newborn calves after nuclear transfer into enucleated oocytes. To date, seven female and six male calves have survived: six of the females were from adult cells (cumulus (n = 3), oviduct (n = 2) and skin (n = 1) cells) and one was from newborn skin cells, whereas the male calves were derived from adult ear cells (n = 3), newborn liver and skin cells (n = 2), and fetal cells (n = 1). Clones derived from adult cells frequently aborted in the later stages of pregnancy and calves developing to term showed a higher number of abnormalities than did those derived from newborn or fetal cells. The telomeric DNA lengths in the ear cells of three male calves cloned from the ear cells of a bull aged 10 years were similar to those of the original bull. However, the telomeric DNA lengths from the white blood cells of the clones, although similar to those in an age-matched control, were shorter than those of the original bull, which indicates that telomeric shortening varies among tissues.
Article
There is much interest in developmentally regulated molecules which may have function in cell interactions and sorting during embryogenesis and differentiation. Numerous antisera have been raised which detect antigens that are expressed in early embryonic cells and become restricted during differentiation, being expressed in only a minority of adult cells (reviewed in refs 1-3). The precise antigenic determinants recognized by such antisera have not been defined. However, studies using a hybri-doma antibody against mouse spleen cells4 and monoclonal autoantibodies of patients with cold agglutinin disease5 have shown that two defined carbohydrate antigen systems, the Forssman and the li antigens, have stage-specific expression in early mouse embryos. We now describe evidence that the stage-specific embryonic antigen SSEA-1 (ref. 6) involves the carbohydrate sequence Galbeta1 --> 4GlcNAc ↑1,3 Fucalpha This determinant is formed by alpha 1 --> 3 fucosylation of blood group I or i antigens which are branched or linear oligosaccharides, respectively7-9, built of Galbeta1 -->4GlcNAc units and known as type 2 precursor chains10 of the major blood group antigens. Thus, we introduce the concept of simple glycosylation changes as a basis for stage-specific expression of embryonic antigens.
Article
The isolation of pluripotent stem cells domestic animals will have a big influence on genetic modification of livestock. Pluripotent stem cell lines have been established for several domestic species. The in vivo pluripotency properties of stem cell lines has been demonstrated for bovine, sheep, and pig stem cell lines. Bovine pluripotent stem cell lines derived from embryos or primordial germ cells are an unlimited source of pluripotent cells. Nuclear transfer of bovine pluripotent stem cells could play an important role in the production of offspring of domestic animals. However, placental development of fetuses derived from nuclear transfer of stem cells is incomplete, several approaches to overcome this problem described. Pluripotent stem cells will become valuable after demonstration of germ line transmission resulting in a full term live calf.
Article
To date, cloned farm animals have been produced by nuclear transfer from embryonic, fetal, and adult cell types. However, mice completely derived from embryonic stem (ES) cells have been produced by aggregation with tetraploid embryos. The objective of the present study was to generate offspring completely derived from bovine ES-like cells. ES-like cells isolated from the inner cell mass of in vitro-produced embryos were aggregated with tetraploid bovine embryos generated by electrofusion at the 2-cell stage. A total of 77 embryo aggregates produced by coculture of two 8-cell-stage tetraploid embryos and a clump of ES-like cells were cultured in vitro. Twenty-eight of the aggregates developed to the blastocyst stage, and 12 of these were transferred to recipient cows. Six calves representing 2 singletons and 2 sets of twins were produced from the transfer of the chimeric embryos. Microsatellite analysis for the 6 calves demonstrated that one calf was chimeric in the hair roots and the another was chimeric in the liver. However, unfortunately, both of these calves died shortly after birth. Two of the placentae from the remaining pregnancies were also chimeric. These results indicate that the bovine ES-like cells used in these studies were able to contribute to development.