Вы находитесь на странице: 1из 9

J. Cell. Mol. Med. Vol 8, No 4, 2004 pp.

465-473

Stem Cell Review Series

Embryonic stem cells:


a promising tool for cell replacement therapy

Michael Xavier Doss, Christoph I. Koehler, C. Gissel,


Jürgen Hescheler, Agapios Sachinidis *

University of Cologne, Center of Physiology and Pathophysiology, Institute of Neurophysiology,


Cologne, Germany

Received: October 26, 2004; Accepted: November 15, 2004

• Introduction – Transplant rejection


– Cell replacement therapy • Advances in CRT with ES cells
• ES cells – Cardiovascular therapy
– Multilineage differentiation in vitro – Haematopoiesis
– ES cells: A powerful source for cell replace- – Therapy for neurological disorders
ment therapy – Treatment of diabetes
• Problems encountered – Liver therapy
– Selection of desired cell phenotype • Future perspectives
– Tumor formation • Conclusion

Abstract

Embryonic stem (ES) cells are revolutionizing the field of developmental biology as a potential tool to understand
the molecular mechanisms occurring during the process of differentiation from the embryonic stage to the adult phe-
notype. ES cells harvested from the inner cell mass (ICM) of the early embryo can proliferate indefinitely in vitro
while retaining the ability to differentiate into all somatic cells. Emerging results from mice models with ES cells are
promising and raising tremendous hope among the scientific community for the ES-cell based cell replacement ther-
apy (CRT) of various severe diseases. ES cells could potentially revolutionize medicine by providing an unlimited
renewable source of cells capable of replacing or repairing tissues that have been damaged in almost all degenera-
tive diseases such as diabetes, myocardial infarction and Parkinson’s disease. This review updates the progress of ES
cell research in CRT, discusses about the problems encountered in the practical utility of ES cells in CRT and eval-
uates how far this approach is successful experimentally.

Keywords: embryonic stem cells • cell replacement therapy

* Correspondence to: Prof. Dr. A. SACHINIDIS Robert Koch Strasse 39, 50931 Cologne, Germany.
University of Cologne, Center of Physiology and Tel.: +49 221 478 73 73, Fax: +49 221 478 6965
Pathophysiology, Institute of Neurophysiology, Email: a.sachinidis@uni-koeln.de
Introduction recombinant proteins such as recombinant insulin
or therapies based on mechanical devices. The
Extensive investigation of embryonic stem (ES) practical utility of this approach mainly depends
cells for the last two decades resulted in the better upon the availability of a versatile source of the
understanding of the molecular processes rightly chosen cells and is being currently delayed
involved during the differentiation process of ES for a long time due to the non-availability and/or
cells to the adult mature phenotype. Under specif- limited supply of a perfect source of cells
ic in vitro culture conditions, ES cells can prolif- required. Much of the research has been drained in
erate indefinitely without senescence and are able order to find out a possible candidate to serve as a
to differentiate into almost all tissue specific cell bank of cells needed for the cell based therapeutic
lineages, if the appropriate extrinsic and intrinsic applications. Several strategies are being currently
stimuli are provided in the culture. These proper- evaluated and include, cell therapies derived from
ties make ES cells an attractive candidate for the autologoues primary cell isolates, cell therapies
so called cell replacement therapy (CRT) of vari- derived from established cell lines, cell therapies
ous degenerative diseases that are associated with derived from a variety of stem cells, including
a loss of functional cells. It is hoped that with the bone marrow and mesenchymal stem cells, cord
ES-based cell replacement therapy of the degen- blood stem cells, ES cells, as well as cells, tissues
erative disorders such as myocardial infarction, and organs from genetically modified animals
diabetes and Parkinson’s disease, normal function [1,2]. The upcoming experimental results with ES
of the respective organs will be restored signifi- cells are encouraging and quite promising [3-6].
cantly. Still, exploitation and practical utility of They give added impetus for this approach.
the ES cells are in their infancy. Further extensive
experimental investigations and clinical trials are
necessary for an optimal ES-based treatment of
degenerative disorders. This review explores how ES cells
well the ES cells are going to be manipulated to
serve as a renewable source of functional cells to ES cells have been in vitro derived in mouse and
be used in the CRT to treat the degenerative dis- human in a similar manner from the inner cell
orders, updates the progress in this regard, dis- mass (ICM) of the blastocysts. In 1981, ES cells
cusses about the problems encountered in the were isolated for the first time in mice and it was
practical utility of the ES cells and outlines how a major breakthrough in the field of developmen-
far this approach will be successful in the near tal biology [7,8]. ES cells can proliferate indefi-
future. nitely without senescence in in vitro culture in an
undifferentiated state in the presence of leukemia
inhibitory factor (LIF) or on top of a layer of
Cell replacement therapy mitotically inactivated mouse embryonic fibrob-
lasts (MEFs) [9]. They can give rise to cell lin-
Degenerative disorders like myocardial infarction, eages of any type of body tissues / organs, when
Parkinson’s disease, diabetes are due to the pro- specific stimuli are provided in the culture (Fig.
gressive (acute/chronic) loss of functional cells 1). The latter property of ES cells is called pluripo-
due to disease or injury to the cells or ageing. tency. ES cells are not totipotent because they fail
Replacing the worn out or injured cells by func- to develop a whole functional organism. To date,
tional cells to restore the normal function of the ES cell lines are available from rodents, rabbits,
tissues or organs is the underlying principle of pigs and primates [7,8,10-12]. Murine ES cell
CRT, otherwise also called as regenerative lines remain undifferentiated when grown either
medicine. It is hoped that the organs or tissues on mitotically inactivated MEFs or in the presence
treated by this approach can perform their normal of relatively high concentrations of leukemia
function more efficiently than the ones treated by inhibitory factor (LIF) whereas human ES cell
conventional therapies like transplantation and lines remain undifferentiated only when grown on
pharmocological therapy like treatment with mitotically inactivated MEFs [13].

466
J. Cell. Mol. Med. Vol 8, No 4, 2004

Fig. 1 Overview of the derivation of ES cells and their pluripotency. ES cells are derived from the ICM of the
early blastocyst and explanted into culture. The ESCA/Embryoid Body is an intermediate stage during in vitro dif-
ferentiation of ES cells into all three different germ layers and their derivates.

mation of ESCAs initiates signalling and sponta-


Multilineage differentiation in vitro neous differentiation of ES cells to the three
embryonic germ layers, the ectoderm, mesoderm
In the absence of LIF or when removed from feed- and endoderm [13,15]. Murine ES cell lines are
er layers and transferred into suspension culture, able to differentiate in vitro into a variety of embry-
ES cells differentiate spontaneously into multicel- onic and adult cell types from all three embryonic
lular ES cell aggregates (ESCAs), which resemble germ layers. These include cardiomyocytes,
early post implantation embryos (Fig. 2) (for hematopoitic progenitors, yolk sac, skeletal
review see [14]). In general, it is believed that for- myoctes, smooth muscle cells, adipocytes, hepato-

467
Fig. 2 In vitro differentiation of ES cells into cardiomyocytes. A: outlines one of the protocols used for generating
ES cell derived cardiomyocytes. B: (I) Es cell derived beating cardiomyocytes in culture. (II) Beating cardiomyocytes
expressing GFP under the control of the cardiac α-actin promotor. (III) and (IV) Immunostaining of the developing
cardiomyocytes in plated EBs with monoclonal antibodies against sarcomeric α-actin. The white arrows indicate beat-
ing cardiac cell clusters. Figure (B II) taken with courtesy of Sachinidis A, Kolossov E, Fleischmann BK, Hescheler J.
Generation of cardiomyocytes from embryonic stem cells experimental studies. Herz. 2002 Nov;27(7):589-97
(Copyright Urban & Vogel. Reproduced with permission). Figure (B III and IV) taken with courtesy of Sachinidis A,
Gissel C, Nierhoff D, Hippler-Altenburg R, Sauer H, Wartenberg M, Hescheler J. Identification of plateled-derived
growth factor-BB as cardiogenesis-inducing factor in mouse embryonic stem cells under serum-free conditions. Cell
Physiol Biochem. 2003;13(6):423-9. (Copyright S. Karger AG, Basel. Reproduced with permission).

cytes, chondrocytes, endothelial cells, melano- phenotype in vitro when appropriate culture condi-
cytes, neurons, glia, pancreatic islet cells, primitive tions are provided and they reflect many of the crit-
endoderm and so on (for review see ref. [15], [16- ical developmental stages found in the normal
27]). These experiments clearly demonstrate that embryo. Figure 2 shows an example for generation
ES cells can be induced to form the desired cell of cardiac cells.

468
J. Cell. Mol. Med. Vol 8, No 4, 2004

ES cells: A powerful source for cell the control of this promoter. Expression of selection
replacement therapy markers (antibiotics resistant genes) makes the
desired population resistant to that particular antibi-
If the appropriate stimuli and/or carefully chosen otic when applied to kill the other cell population.
combination of extrinsic and intrinsic signal fac- These antibiotics block the endogenous translation
tors are available in culture, ES cells will differen- or transcription machineries if cells do not express
tiate into almost all cell lineages of body tissues the particular enzyme/ protein which antagonizes
like cardiomyocytes, oligodendrocytes, astrocytes, the blocking activity of these antibiotics.
beta cells of the pancreas, hepatic cells and so on Expression of the fluorescent marker allows the
[28]. Several transcription factors have been desired cell phenotype to be harvested by FACS.
demonstrated to regulate differentiation of ES The drawback of these two approaches is that the
cells to specific cell types [29]. Ectopic overex- genetically manipulated ES cell derivatives might
pression of such factors stimulates ES cells to dif- be immunologically rejected since they express the
ferentiate selectively into certain cell types [30- foreign proteins -the selection and the fluorescent
32]. Also several other substances like ascorbic markers.
acid, butyric acid and DMSO enhance the differ-
entiation of ES cells to specific mature phenotypes
[33,34]. Tumor formation

Implantation of undifferentiated ES cells leads to


formation of benign teratoma in the recipients
Problems encountered [12,13]. This demands a pure population of termi-
nally differentiated cell phenotype. Negative selec-
Selection of desired cell phenotype tion of Oct4 expressing cells might be a solution for
this issue. New strategies and methodologies need
The routinely used differentiation protocols give to be developed to isolate the terminally differenti-
rise to a mixture of different cell populations.To ated cells. ES cell implants can be tagged with some
harvest a single population of cells of our interest, kind of death signals in such a way that when they
several strategies have been developed. Magnetic start to form tumors by accident, or when they start
bead tagged antibodies directed against the unique to cause severe complications, they will be cleared
cell surface marker of the desired cell population off from the body leaving the host unaffected. New
can be directed and the desired population can be methodologies and the forthcoming new discover-
harvested by running through a magnetic column ies will find a possible way for this approach.
where the magnet tagged antibodies bound with the
desired cell popuation will be retained, allowing
other cell population washed out. Fluorescence Transplant rejection
labelled antibodies raised against unique cell sur-
face markers of the desired cell population can be ES cell derived cellular grafts might be rejected due
lifted up by the fluorescence -activated cell sorting to immunogenicity of the transplanted cells. ES
(FACS). Some protocols employ selective culture cells can be easily manipulated genetically to over-
conditions that promote the growth of one particu- come the problem of immune rejection so that life-
lar cell type at the expense of the other. ES cells long pharmocologic immunosuppression will not
may be genetically engineered to have selection be needed. One attractive approach is that the MHC
markers or fluorescent markers under the control of genes in ES cells can be replaced with host MHC
a lineage/tissue specific promoter, for example a genes so that the host immune system will recog-
transcription factor that is switched on early during nise the implanted cells as ‘self’ [35,36]. This
lineage specific differentiation. When the tissue/lin- approach is practically possible with help of homol-
eage specific promoter is activated during the par- ogous recombination techniques. An alternative
ticular lineage differentiation, the selection marker approach will be the insertion of genes for the
or the fluorescent marker will be expressed under immunosuppressive molecules such as Fas- ligand

469
into the ES cells [37,38]. Additionally the immuno- steadily, necessitating the vital need for the devel-
logically-reactive molecules such as B7 antigens, or opment of new therapeutic options. In the last
CD40 ligand present on the surface of the ES cells decade, several animal studies demonstrated that
could be abrogated from ES cells [37,38]. One transplantation of isolated cardiomyocytes might
interesting approach in this regard is the application offer an alternative approach for the treatment of
of nuclear transplantation technology in ES cells. In cardiac degenerative disorders. Several animal
this approach, the nucleus from a normal somatic studies demonstrate that engraftment of cardiac
cell of the recipient, say from a skin biopsy is myocytes into the adult heart can be achieved suc-
extracted and is then injected into an enucleated cessfully. The emerging results with ES cells have
oocyte. The cytoplasm of this oocyte has the the potential to revolutionize Medicine and could
required potential to reprogram the differentiated find a cure for the cardiac degenerative disorders
nucleus injected and re-establishes an embryonic obviating the need for the allogeneic organ trans-
gene expression pattern in the chromatin of the plantation. Several factors alone or in combination
somatic cell nucleus. The blastocyst formed from have been shown to enrich cardiac differentiation
this oocyte would be the source for the derivation of such as hepatocyte growth factor (HGF), epider-
new ES cell lines which would be genetically mal growth factor (EGF), basic fibroblast growth
matched for each nuclear gene of the recipient factor (bFGF), transforming growth factor b1
[39,40]. In this case, whole MHC regions in ES (TGFb1), platelet derived growth factor (PDGF),
cells and other immune molecules will be identical sphingosine-1-phosphate, retinoic acid, 5-azacyti-
to that of the patient, except minor molecules dine, vitamin C, and overexpression of GATA-4
derived from mitochondrial genes. The immune (For review see [41], [28,30,33,42,43] The so
rejection in this case will be the least of all the called CRT where the damaged cardiac cells will
above-mentioned approaches. However, although be replaced by cells derived from the ES cells to
the therapeutical cloning approach appears to be restore the normal function of the heart could be a
promising for avoiding possible immunological boon for heart patients in near future.
problems by the recipients, this approach bears sev-
eral ethical problems that should be solved by
social consensus before clinical studies in this Haematopoiesis
direction can be initiated.
Hematopoiesis in ESCAs develops spontaneous-
ly in the absence of exogenous cytokines demon-
strating that the microenvironment of the devel-
Advances in CRT with ES cells oping ESCAs is enough to supply the necessary
inductive signals for commitment of mesodermal
Despite the pitfalls discussed above, there are precursors to haematopoietic cell fate, and also to
more interesting results coming out with ES cells provide the differentiation signals for blood cell
for the therapy of several degenerative disorders maturation. The pattern of haematopoiesis in the
as follows. ESCAs mimics yolk sac development and is
dominated by primitive erythroid cells that
express embryonic forms of hemoglobin (bH1)
Cardiovascular therapy [21,44].

Several cardiovascular diseases are associated


with myocardial cell death resulting from Therapy for neurological disorders
ischemic heart disease, viral infections or
immunopathological conditions. The only current Neuroectodermal differentiation is also relatively
treatment for the patients with end stage heart fail- easy to achieve from murine ES cells. High yields
ure is the cardiac transplantation. Often, this is of neurons, astrocytes and oligodendrocyte like
limited by the availability of a perfectly matched cells have been achieved with several different
organ. The demand for organ supply is increasing optimised protocols. These protocols have since

470
J. Cell. Mol. Med. Vol 8, No 4, 2004

been further simplified to remove the ESCAs Future perspectives


stage and differentiate murine and human ES cells
into neuroectodermal lineages in monolayer. This To serve as a versatile renewable source of
is the only somatic cell type for which this is healthier cells for the use in CRT, the problems
known to be possible. Preliminary implantation mentioned above should be overcome. That is,
studies of ES derived neurons into mouse models new methodologies and protocols should be
have reported that these cells are functionally developed for the derivation of every single cell
active, integrate into the brain and in one case lineage population of body tissues/organs. New
have corrected the phenotype of a neurodegenera- strategies should be developed to harvest only
tive disease [45,45-47]. the terminally differentiated phenotype of the
cell or the phenotype of the cell which serves as
nearly an ideal substitute to be used for CRT.
Treatment of diabetes This population of cells should be free from the
tumourigenic population or from the population
β-cell replacement therapy via islet transplantation that may lead to severe complications during the
through stem cells has gained more popularity due post transplantation period. The problem of the
to the recent upcoming results in the animal model immnogenicity of ES cell derived cellular trans-
studies. Early investigations demonstrate that ES plant in the host should be overcome by new
cells are the promising sources for alleviating the approaches. Once all these issues will be
pathological conditions of diabetic patients [48]. It addressed and a proper solution is found to over-
has been reported that the insulin-expressing cells come these obstacles and hurdles, ES cells will
that had spontaneously differentiated within prove to be the potential versatile source of the
embryoid bodies were selected by an approach cellular transplants to be used in CRT to treat a
where an introduced antibiotic resistance marker number of degenerative disorders. The upcoming
under the control of the insulin promoter was new discoveries and better understanding of the
made use of [49]. molecular processes involved during the transi-
tion of embryonic stage to the adult phenotype
are opening up new avenues to overcome the
Liver therapy above mentioned hurdles and they will convert
our hopes into reality in the near future.
Hepatocytes or hepatocyte-like cells derived from
ES cells can be a promising approach in the therapy
of liver diseases. Several entities like chronic viral
hepatitis, alcoholic liver disease, haemochromatosis Conclusion
etc. result in liver cirrhosis and finally organ failure
making a liver transplantation inevitable. Utilization and practical application of ES cells in
Differentiated ES cells could be used either for direct cell replacement therapy are still in their infancy
transplantation into the diseased liver or for the gen- and need further exploration and extensive inves-
eration of bioartificial liver (BAL) devices to bypass tigations, experimental confirmations and clinical
situations of acute liver failure until an organ for trials before they will be made available as an
transplantation is available, for example acute fulmi- ideal cell substitute for the treatment of a number
nant viral hepatitis or severe intoxications. Studies of degenerative disorders. The daily upcoming
have shown the differentiation of murine and human experimental observations are reinforcing the
ES cells into hepatocyte-like cells and results have solid hope that ES cells will be the potential
also been achieved on the transplantation of differen- source for use in cell replacement therapy. Once
tiated murine ES cell into the liver [50-54]. first successful clinical studies will be achieved
Especially BAL devices seem to be promising for the and evaluated, ES-based cell replacement therapy
next future. Main difficulties of CRT like tumor for- will revolutionize medicine in the near future
mation or problematic engraftment of the cells are of offering therapeutical alternatives for treatment of
no relevance in this therapeutic approach. severe degenerative disorders.

471
Differentiation of human embryonic stem cells into
References embryoid bodies compromising the three embryonic
germ layers, Mol. Med. 6: 88-95, 2000
1. Asahara T., Kalka C., Isner J.M., Stem cell therapy and 16. Odorico J.S., Kaufman D.S., Thomson J.A.,
gene transfer for regeneration, Gene Ther., 7: 451-457, Multilineage differentiation from human embryonic stem
2000 cell lines, Stem Cells 19: 193-204, 2001
2. Fodor W.L., Tissue engineering and cell based therapies, 17. Wobus A.M., Grosse R., Schoneich J., Specific effects
from the bench to the clinic: the potential to replace, of nerve growth factor on the differentiation pattern of
repair and regenerate, Reprod. Biol. Endocrinol. 1: 102, mouse embryonic stem cells in vitro, Biomed. Biochim.
2003 Acta 47: 965-973, 1988
3. Delcarpio J.B., Claycomb W.C., Cardiomyocyte trans- 18. Rathjen P.D., Lake J., Whyatt L.M., Bettess M.D.,
fer into the mammalian heart. Cell-to-cell interactions in Rathjen J., Properties and uses of embryonic stem cells:
vivo and in vitro, Ann. N. Y. Acad. Sci. 752: 267-285, prospects for application to human biology and gene
1995 therapy, Reprod. Fertil. Dev. 10: 31-47, 1998
4. Koh G.Y., Soonpaa M.H., Klug M.G., Field L.J., Long- 19. Doetschman T.C., Eistetter H., Katz M., Schmidt W.,
term survival of AT-1 cardiomyocyte grafts in syngeneic Kemler R., The in vitro development of blastocyst-
myocardium, Am. J. Physiol. 264: H1727-H1733, 1993 derived embryonic stem cell lines: formation of visceral
5. Koh G.Y., Soonpaa M.H., Klug M.G., Pride H.P., yolk sac, blood islands and myocardium, J. Embryol.
Cooper B.J., Zipes D.P., Field L.J., Stable fetal car- Exp. Morphol. 87: 27-45, 1985
diomyocyte grafts in the hearts of dystrophic mice and 20. Baker R.K., Lyons G.E., Embryonic stem cells and in
dogs, J. Clin. Invest. 96: 2034-2042, 1995 vitro muscle development, Curr. Top. Dev. Biol. 33: 263-
6. Roell W., Lu Z.J., Bloch W., Siedner S., Tiemann K., 279, 1996
Xia Y., Stoecker E., Fleischmann M., Bohlen H., 21. Keller G., Kennedy M., Papayannopoulou T., Wiles
Stehle R., Kolossov E., Brem G., Addicks K., Pfitzer M.V., Hematopoietic commitment during embryonic
G., Welz A., Hescheler J., Fleischmann B.K., Cellular stem cell differentiation in culture, Mol. Cell Biol. 13:
cardiomyoplasty improves survival after myocardial 473-486, 1993
injury, Circulation 105: 2435-2441, 2002 22. Rohwedel J., Maltsev V., Bober E., Arnold H.H.,
7. Evans M.J., Potential for genetic manipulation of mam- Hescheler J., Wobus A.M., Muscle cell differentiation
mals, Mol. Biol. Med. 6: 557-565, 1989 of embryonic stem cells reflects myogenesis in vivo:
8. Martin G.R., Isolation of a pluripotent cell line from developmentally regulated expression of myogenic
early mouse embryos cultured in medium conditioned by determination genes and functional expression of ionic
teratocarcinoma stem cells, Proc. Natl. Acad. Sci. USA currents, Dev. Biol. 164: 87-101, 1994
78: 7634-7638, 1981 23. Dani C., Smith A.G., Dessolin S., Leroy P., Staccini L.,
9. Williams R.L., Hilton D.J., Pease S., Willson T.A., Villageois P., Darimont C., Ailhaud G., Differentiation
Stewart C.L., Gearing D.P., Wagner E.F., Metcalf D., of embryonic stem cells into adipocytes in vitro, J. Cell
Nicola N.A., Gough N.M., Myeloid leukaemia inhibito- Sci. 110: 1279-1285, 1997
ry factor maintains the developmental potential of 24. Poliard A., Nifuji A., Lamblin D., Plee E., Forest C.,
embryonic stem cells, Nature 336: 684-687, 1988 Kellermann O., Controlled conversion of an immortal-
10. Graves K.H., Moreadith R.W., Derivation and charac- ized mesodermal progenitor cell towards osteogenic,
terization of putative pluripotential embryonic stem cells chondrogenic, or adipogenic pathways, J. Cell Biol. 130:
from preimplantation rabbit embryos, Mol. Reprod. Dev. 1461-1472, 1995
36: 424-433, 1993 25. Risau W., Sariola H., Zerwes H.G., Sasse J., Ekblom
11. Li M., Zhang D., Hou Y., Jiao L., Zheng X., Wang P., Kemler R., Doetschman T., Vasculogenesis and
W.H., Isolation and culture of embryonic stem cells from angiogenesis in embryonic-stem-cell-derived embryoid
porcine blastocysts, Mol. Reprod. Dev. 65: 429-434, bodies, Development 102: 471-478, 1988
2003 26. Yamane T., Hayashi S., Mizoguchi M., Yamazaki H.,
12. Thomson J.A., Kalishman J., Golos T.G., Durning M., Kunisada T., Derivation of melanocytes from embryon-
Harris C.P., Becker R.A., Hearn J.P., Isolation of a pri- ic stem cells in culture, Dev. Dyn. 216: 450-458, 1999
mate embryonic stem cell line, Proc. Natl. Acad. Sci. 27. Brustle O., Jones K.N., Learish R.D., Karram K.,
USA 92: 7844-7848, 1995 Choudhary K., Wiestler O.D., Duncan I.D., McKay
13. Thomson J.A., Itskovitz-Eldor J., Shapiro S.S., R.D., Embryonic stem cell-derived glial precursors: a
Waknitz M.A., Swiergiel J.J., Marshall V.S., Jones source of myelinating transplants, Science 285: 754-756,
J.M., Embryonic stem cell lines derived from human 1999
blastocysts, Science 282: 1145-1147, 1998 28. Sachinidis A., Gissel C., Nierhoff D., Hippler-
14. Sachinidis A., Fleischmann B.K., Kolossov E., Altenburg R., Sauer H., Wartenberg M., Hescheler J.,
Wartenberg M., Sauer H., Hescheler J., Cardiac spe- Identification of plateled-derived growth factor-BB as
cific differentiation of mouse embryonic stem cells, cardiogenesis-inducing factor in mouse embryonic stem
Cardiovasc. Res. 58: 278-291, 2003 cells under serum-free conditions, Cell Physiol Biochem.
15. Itskovitz-Eldor J., Schuldiner M., Karsenti D., Eden 13: 423-429, 2003
A., Yanuka O., Amit M., Soreq H., Benvenisty N.,

472
J. Cell. Mol. Med. Vol 8, No 4, 2004

29. Levinson-Dushnik M., Benvenisty N., Involvement of the differentiation of cells derived from human embryon-
hepatocyte nuclear factor 3 in endoderm differentiation ic stem cells, Proc. Natl. Acad. Sci. U. S. A 97: 11307-
of embryonic stem cells, Mol. Cell Biol. 17: 3817-3822, 11312, 2000
1997 43. Xu C., Police S., Rao N., Carpenter M.K., Characte-
30. Grepin C., Nemer G., Nemer M., Enhanced cardiogen- rization and enrichment of cardiomyocytes derived from
esis in embryonic stem cells overexpressing the GATA-4 human embryonic stem cells, Circ. Res. 91: 501-508,
transcription factor, Development 124: 2387-2395, 1997 2002
31. Fujikura J., Yamato E., Yonemura S., Hosoda K., 44. Kennedy M., Firpo M., Choi K., Wall C., Robertson
Masui S., Nakao K., Miyazaki J.J., Niwa H., S., Kabrun N., Keller G., A common precursor for prim-
Differentiation of embryonic stem cells is induced by itive erythropoiesis and definitive haematopoiesis,
GATA factors, Genes Dev. 16: 784-789, 2002 Nature 386: 488-493, 1997
32. Kanda S., Shiroi A., Ouji Y., Birumachi J., Ueda S., 45. Rippon H.J., Bishop A.E., Embryonic stem cells, Cell
Fukui H., Tatsumi K., Ishizaka S., Takahashi Y., Prolif. 37: 23-34, 2004
Yoshikawa M., In vitro differentiation of hepatocyte-like 46. Barberi T., Klivenyi P., Calingasan N.Y., Lee H.,
cells from embryonic stem cells promoted by gene trans- Kawamata H., Loonam K., Perrier A.L., Bruses J.,
fer of hepatocyte nuclear factor 3 beta, Hepatol. Res. 26: Rubio M.E., Topf N., Tabar V., Harrison N.L., Beal
225-231, 2003 M.F., Moore M.A., Studer L., Neural subtype specifica-
33. Takahashi T., Lord B., Schulze P.C., Fryer R.M., tion of fertilization and nuclear transfer embryonic stem
Sarang S.S., Gullans S.R., Lee R.T., Ascorbic acid cells and application in parkinsonian mice, Nat.
enhances differentiation of embryonic stem cells into Biotechnol. 21: 1200-1207, 2003
cardiac myocytes, Circulation 107: 1912-1916, 2003 47. Chiba S., Iwasaki Y., Sekino H., Suzuki N.,
34. Ventura C., Maioli M., Asara Y., Santoni D., Scarlata Transplantation of motoneuron-enriched neural cells
I., Cantoni S., Perbellini A., Butyric and retinoic mixed derived from mouse embryonic stem cells improves
ester of hyaluronan. A novel differentiating glycoconju- motor function of hemiplegic mice, Cell Transplant. 12:
gate affording a high throughput of cardiogenesis in 457-468, 2003
embryonic stem cells, J. Biol. Chem. 279: 23574-23579, 48. Bonner-Weir S., Stem cells in diabetes: what has been
2004 achieved, Horm. Res. 60 Suppl 3: 10, 2003
35. Hardy R.R., Malissen B., Lymphocyte development. 49. Soria B., Skoudy A., Martin F., From stem cells to beta
The (knock-) ins and outs of lymphoid development, cells: new strategies in cell therapy of diabetes mellitus,
Curr. Opin. Immunol. 10: 155-157, 1998 Diabetologia 44: 407-415, 2001
36. Westphal C.H., Leder P., Transposon-generated 50. Jones E.A., Tosh D., Wilson D.I., Lindsay S.,
‘knock-out’ and ‘knock-in’ gene-targeting constructs for Forrester L.M., Hepatic differentiation of murine
use in mice, Curr. Biol. 7: 530-533, 1997 embryonic stem cells, Exp. Cell Res. 272: 15-22, 2002
37. Harlan D.M., Kirk A.D., The future of organ and tissue 51. Rambhatla L., Chiu C.P., Kundu P., Peng Y.,
transplantation: can T-cell costimulatory pathway modi- Carpenter M.K., Generation of hepatocyte-like cells
fiers revolutionize the prevention of graft rejection?, from human embryonic stem cells, Cell Transplant. 12:
JAMA 282: 1076-1082, 1999 1-11, 2003
38. Walker P.R., Saas P., Dietrich P.Y., Role of Fas ligand 52. Hamazaki T., Iiboshi Y., Oka M., Papst P.J.,
(CD95L) in immune escape: the tumor cell strikes back, Meacham A.M., Zon L.I., Terada N., Hepatic matura-
J. Immunol. 158: 4521-4524, 1997 tion in differentiating embryonic stem cells in vitro,
39. Hochedlinger K., Jaenisch R., Nuclear transplantation, FEBS Lett. 497: 15-19, 2001
embryonic stem cells, and the potential for cell therapy, 53. Yamamoto H., Quinn G., Asari A., Yamanokuchi H.,
N. Engl. J. Med. 349: 275-286, 2003 Teratani T., Terada M., Ochiya T., Differentiation of
40. Hochedlinger K., Rideout W.M., Kyba M., Daley embryonic stem cells into hepatocytes: biological func-
G.Q., Blelloch R., Jaenisch R., Nuclear transplantation, tions and therapeutic application, Hepatology 37: 983-
embryonic stem cells and the potential for cell therapy, 993, 2003
Hematol. J. 5 Suppl 3: S114-S117, 2004 54. Yamada T., Yoshikawa M., Kanda S., Kato Y.,
41. Lovell M.J., Mathur A., The role of stem cells for treat- Nakajima Y., Ishizaka S., Tsunoda Y., In vitro differen-
ment of cardiovascular disease, Cell Prolif. 37: 67-87, 2004 tiation of embryonic stem cells into hepatocyte-like cells
42. Schuldiner M., Yanuka O., Itskovitz-Eldor J., Melton identified by cellular uptake of indocyanine green, Stem
D.A., Benvenisty N., Effects of eight growth factors on Cells 20: 146-154, 2002

473

Вам также может понравиться